1
|
Zhang X, Lee JY, Pacheco J, Sutkeviciute I, Anitha AK, Liu H, Singh S, Ventura C, Savransky S, Khatri A, Zhang C, Bahar I, Vilardaga JP. Allosteric mechanism in the distinctive coupling of G q and G s to the parathyroid hormone type 1 receptor. Proc Natl Acad Sci U S A 2025; 122:e2426178122. [PMID: 40138341 PMCID: PMC12002267 DOI: 10.1073/pnas.2426178122] [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] [Received: 12/13/2024] [Accepted: 02/13/2025] [Indexed: 03/29/2025] Open
Abstract
The mechanism determining the preferential stimulation of one heterotrimeric G protein signaling pathway over another by a ligand remains undetermined. By reporting the cryogenic electron microscopy (cryo-EM) structure of the parathyroid hormone (PTH) type 1 receptor (PTH1R) complexed with Gq and comparing its allosteric dynamics with that of PTH1R in complex with Gs, we uncover a mechanism underlying such preferences. We show that an allosteric coupling between the ligand PTH and the C-terminal helix α5 of the Gα subunit controls the stability of the PTH1R complex with the specific G protein, Gs or Gq. Single-cell-level experiments further validate the G protein-selective effects of the PTH binding pose by demonstrating the differential, G protein-dependent residence times and affinity of this ligand at the PTH1R binding site. The findings deepen our understanding of the selective coupling of PTH1R to Gs or Gq and how it relates to the stability and kinetics of ligand binding. They explain the observed variability in the ligand-binding affinity of a GPCR when coupled to different G proteins.
Collapse
MESH Headings
- Receptor, Parathyroid Hormone, Type 1/metabolism
- Receptor, Parathyroid Hormone, Type 1/chemistry
- Receptor, Parathyroid Hormone, Type 1/genetics
- GTP-Binding Protein alpha Subunits, Gq-G11/metabolism
- GTP-Binding Protein alpha Subunits, Gq-G11/chemistry
- Humans
- Allosteric Regulation
- Parathyroid Hormone/metabolism
- Parathyroid Hormone/chemistry
- GTP-Binding Protein alpha Subunits, Gs/metabolism
- GTP-Binding Protein alpha Subunits, Gs/chemistry
- Protein Binding
- Cryoelectron Microscopy
- Ligands
- HEK293 Cells
- Binding Sites
- Signal Transduction
- Kinetics
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA15261
| | - Ji Young Lee
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, New York, NY11794
- Department of Biochemistry and Cell Biology, Renaissance School of Medicine, Stony Brook University, New York, NY11794
| | - Jonathan Pacheco
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA15261
| | - Ieva Sutkeviciute
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA15261
| | - Anju Krishnan Anitha
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Heng Liu
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA15261
| | - Stephanie Singh
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA15261
| | - Carlos Ventura
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, New York, NY11794
- Department of Biochemistry and Cell Biology, Renaissance School of Medicine, Stony Brook University, New York, NY11794
| | - Sofya Savransky
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA15261
| | - Ashok Khatri
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Cheng Zhang
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA15261
| | - Ivet Bahar
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, New York, NY11794
- Department of Biochemistry and Cell Biology, Renaissance School of Medicine, Stony Brook University, New York, NY11794
| | - Jean-Pierre Vilardaga
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA15261
- United States Department of Veterans Affairs, Pittsburgh Veterans Affairs Health Care System, Pittsburgh, PA15240
| |
Collapse
|
2
|
Zhang Y, Zheng Q, Warshel A, Bai C. Key Interaction Changes Determine the Activation Process of Human Parathyroid Hormone Type 1 Receptor. J Am Chem Soc 2025; 147:3539-3552. [PMID: 39804793 DOI: 10.1021/jacs.4c15025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The parathyroid hormone type 1 receptor (PTH1R) plays a crucial role in modulating various physiological functions and is considered an effective therapeutic target for osteoporosis. However, a lack of detailed molecular and energetic information about PTH1R limits our comprehensive understanding of its activation process. In this study, we performed computational simulations to explore key events in the activation process, such as conformational changes in PTH1R, Gs protein coupling, and the release of guanosine diphosphate (GDP). Our analysis identified kinetic information, including the rate-determining step, transition state, and energy barriers. Free-energy and structural analyses revealed that GDP could be released from the Gs protein when the binding cavity is partially open. Additionally, we predicted important residues, including potential pathogenic mutations, and verified their significance through site-directed mutations. These findings enhance our understanding of class B GPCR activation mechanisms. Furthermore, the methodology employed in this study can be applied to other biophysical systems.
Collapse
Affiliation(s)
- Yue Zhang
- School of Chemistry and Environmental Engineering, Changchun University of Science and Technology, Changchun 130012, China
- Warshel Institute for Computational Biology, School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong (Shenzhen), Shenzhen 518172, China
| | - Qingchuan Zheng
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China
| | - Arieh Warshel
- Department of Chemistry, University of Southern California, Los Angeles, California 90089-1062, United States
| | - Chen Bai
- Warshel Institute for Computational Biology, School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong (Shenzhen), Shenzhen 518172, China
- Chenzhu (MoMeD) Biotechnology Co., Ltd., Hangzhou 310005, China
| |
Collapse
|
3
|
Modafferi C, Tabolacci E, Grippaudo C, Chiurazzi P. Syndromic and Non-Syndromic Primary Failure of Tooth Eruption: A Genetic Overview. Genes (Basel) 2025; 16:147. [PMID: 40004475 PMCID: PMC11855040 DOI: 10.3390/genes16020147] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Primary failure of tooth eruption (PFE) is a rare genetic disorder characterized by the failure of teeth to erupt in the absence of obvious physical obstructions, often resulting in a progressive open bite that is resistant to orthodontic treatment. While PFE can be caused by genetic or systemic factors (such as cysts, tumors, and endocrine imbalances), the non-syndromic causes are primarily genetic, with an autosomal dominant inheritance pattern with variable expressivity. Several genes have been closely associated with the non-syndromic PFE form. The PTH1R (parathyroid hormone 1 receptor) is the most commonly PFE-associated gene. Additional genes associated with minor frequency are Transmembrane protein 119 (TMEM119), which reduces the glycolytic efficiency of bone cells, limiting their mineralization capacity and causing bone fragility; Periostin (POSTN), which regulates the extracellular matrix and the bone's response to mechanical stress; and Lysine (K)-specific methyltransferase 2C (KMT2C), which establishes histone methylation near the Wnt Family Member 5A (WNT5A) gene involved in dental development (odontogenesis). Syndromic forms of PFE are typically associated with complex multisystem disorders, where dental eruption failure is one of the clinical features of the spectrum. These syndromes are often linked to genetic variants that affect ectodermal development, craniofacial patterning, and skeletal growth, leading to abnormal tooth development and eruption patterns. Notable syndromes include GAPO syndrome, ectodermal dysplasia, and cleidocranial dysplasia, each contributing to PFE through distinct molecular mechanisms, such as disruptions in dental structure development, cranial abnormalities, or systemic developmental delays. The main aim of this review is to provide a comprehensive overview of the genetic basis underlying both syndromic and non-syndromic forms of PFE to facilitate precision diagnosis, foster the development of personalized therapeutic strategies, and offer new insights into managing this complex dental anomaly.
Collapse
Affiliation(s)
- Clarissa Modafferi
- UOC Genetica Medica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (C.M.); (P.C.)
| | - Elisabetta Tabolacci
- Dipartimento di Scienze della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Cristina Grippaudo
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
- UOC Clininica Odontoiatrica, Dipartimento di Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
- Odontoiatria e Protesi Dentaria, Dipartimento Testa Collo ed Organi di Senso, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Pietro Chiurazzi
- UOC Genetica Medica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (C.M.); (P.C.)
- Dipartimento di Scienze della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| |
Collapse
|
4
|
Walker V. The Molecular Biology of Placental Transport of Calcium to the Human Foetus. Int J Mol Sci 2025; 26:383. [PMID: 39796238 PMCID: PMC11720126 DOI: 10.3390/ijms26010383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
From fertilisation to delivery, calcium must be transported into and within the foetoplacental unit for intracellular signalling. This requires very rapid, precisely located Ca2+ transfers. In addition, from around the eighth week of gestation, increasing amounts of calcium must be routed directly from maternal blood to the foetus for bone mineralisation through a flow-through system, which does not impact the intracellular Ca2+ concentration. These different processes are mediated by numerous membrane-sited Ca2+ channels, transporters, and exchangers. Understanding the mechanisms is essential to direct interventions to optimise foetal development and postnatal bone health and to protect the mother and foetus from pre-eclampsia. Ethical issues limit the availability of human foetal tissue for study. Our insight into the processes of placental Ca2+ handling is advancing rapidly, enabled by developing genetic, analytical, and computer technology. Because of their diverse sources, the reports of new findings are scattered. This review aims to pull the data together and to highlight areas of uncertainty. Areas needing clarification include trafficking, membrane expression, and recycling of channels and transporters in the placental microvilli; placental metabolism of vitamin D in gestational diabetes and pre-eclampsia; and the vascular effects of increased endothelial Orai expression by pregnancy-specific beta-1-glycoproteins PSG1 and PSG9.
Collapse
Affiliation(s)
- Valerie Walker
- Department of Clinical Biochemistry, University Hospital Southampton NHS Foundation Trust, Southampton General Hospital, Southampton SO16 6YD, UK
| |
Collapse
|
5
|
Schöneberg T. Modulating vertebrate physiology by genomic fine-tuning of GPCR functions. Physiol Rev 2025; 105:383-439. [PMID: 39052017 DOI: 10.1152/physrev.00017.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/08/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024] Open
Abstract
G protein-coupled receptors (GPCRs) play a crucial role as membrane receptors, facilitating the communication of eukaryotic species with their environment and regulating cellular and organ interactions. Consequently, GPCRs hold immense potential in contributing to adaptation to ecological niches and responding to environmental shifts. Comparative analyses of vertebrate genomes reveal patterns of GPCR gene loss, expansion, and signatures of selection. Integrating these genomic data with insights from functional analyses of gene variants enables the interpretation of genotype-phenotype correlations. This review underscores the involvement of GPCRs in adaptive processes, presenting numerous examples of how alterations in GPCR functionality influence vertebrate physiology or, conversely, how environmental changes impact GPCR functions. The findings demonstrate that modifications in GPCR function contribute to adapting to aquatic, arid, and nocturnal habitats, influencing camouflage strategies, and specializing in particular dietary preferences. Furthermore, the adaptability of GPCR functions provides an effective mechanism in facilitating past, recent, or ongoing adaptations in animal domestication and human evolution and should be considered in therapeutic strategies and drug development.
Collapse
Affiliation(s)
- Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
- School of Medicine, University of Global Health Equity, Kigali, Rwanda
| |
Collapse
|
6
|
Fan Y, Lyu P, Wang J, Wei Y, Li Z, Zhang S, Ouchi T, Jing J, Yuan Q, Rosen CJ, Zhou C. Negative feedback between PTH1R and IGF1 through the Hedgehog pathway in mediating craniofacial bone remodeling. JCI Insight 2024; 10:e183684. [PMID: 39688917 PMCID: PMC11948590 DOI: 10.1172/jci.insight.183684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024] Open
Abstract
Regeneration of orofacial bone defects caused by inflammation-related diseases or trauma remains an unmet challenge. Parathyroid hormone 1 receptor (PTH1R) signaling is a key mediator of bone remodeling whereas the regulatory mechanisms of PTH1R signaling in oral bone under homeostatic or inflammatory conditions have not been demonstrated by direct genetic evidence. Here, we observed that deletion of PTH1R in Gli1+ progenitors led to increased osteogenesis and osteoclastogenesis. Single-cell and bulk RNA-Seq analysis revealed that PTH1R suppressed the osteogenic potential of Gli1+ progenitors during inflammation. Moreover, we identified upregulated IGF1 expression upon PTH1R deletion. Dual deletion of IGF1 and PTH1R ameliorated the bone-remodeling phenotypes in PTH1R-deficient mice. Furthermore, in vivo evidence revealed an inverse relationship between PTH1R and Hedgehog signaling, which was responsible for the upregulated IGF1 production. Our work underscored the negative feedback between PTH1R and IGF1 in craniofacial bone turnover and revealed mechanisms modulating orofacial bone remodeling.
Collapse
Affiliation(s)
- Yi Fan
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Department of Cariology and Endodontics
| | - Ping Lyu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Department of Cariology and Endodontics
| | - Jiahe Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Department of Pediatric Dentistry, and
| | - Yali Wei
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Department of Cariology and Endodontics
| | - Zucen Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Department of Cariology and Endodontics
| | - Shiwen Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Takehito Ouchi
- Department of Physiology, Tokyo Dental College, Tokyo, Japan
| | - Junjun Jing
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | | | - Chenchen Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases
- Department of Pediatric Dentistry, and
| |
Collapse
|
7
|
Hernández-García F, Fernández-Iglesias Á, Rodríguez Suárez J, Gil Peña H, López JM, Pérez RF. The Crosstalk Between Cartilage and Bone in Skeletal Growth. Biomedicines 2024; 12:2662. [PMID: 39767569 PMCID: PMC11727353 DOI: 10.3390/biomedicines12122662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 01/04/2025] Open
Abstract
While the flat bones of the face, most of the cranial bones, and the clavicles are formed directly from sheets of undifferentiated mesenchymal cells, most bones in the human body are first formed as cartilage templates. Cartilage is subsequently replaced by bone via a very tightly regulated process termed endochondral ossification, which is led by chondrocytes of the growth plate (GP). This process requires continuous communication between chondrocytes and invading cell populations, including osteoblasts, osteoclasts, and vascular cells. A deeper understanding of these signaling pathways is crucial not only for normal skeletal growth and maturation but also for their potential relevance to pathophysiological processes in bones and joints. Due to limited information on the communication between chondrocytes and other cell types in developing bones, this review examines the current knowledge of how interactions between chondrocytes and bone-forming cells modulate bone growth.
Collapse
Affiliation(s)
- Frank Hernández-García
- Departamento de Medicina, Oviedo University, 33003 Oviedo, Spain; (F.H.-G.); (J.R.S.)
- Grupo Investigación Pediatría, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (Á.F.-I.); (H.G.P.); (J.M.L.)
| | - Ángela Fernández-Iglesias
- Grupo Investigación Pediatría, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (Á.F.-I.); (H.G.P.); (J.M.L.)
| | - Julián Rodríguez Suárez
- Departamento de Medicina, Oviedo University, 33003 Oviedo, Spain; (F.H.-G.); (J.R.S.)
- Grupo Investigación Pediatría, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (Á.F.-I.); (H.G.P.); (J.M.L.)
- AGC de Infancia y Adolescencia, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- RICORS-SAMID (RD21/0012), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Helena Gil Peña
- Grupo Investigación Pediatría, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (Á.F.-I.); (H.G.P.); (J.M.L.)
- AGC de Infancia y Adolescencia, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- RICORS2040 (RD21/0005/0011), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José M. López
- Grupo Investigación Pediatría, Instituto de Investigación Sanitaria del Principado de Asturias, 33011 Oviedo, Spain; (Á.F.-I.); (H.G.P.); (J.M.L.)
- Departamento de Morfología y Biología Celular, Oviedo University, 33003 Oviedo, Spain
| | - Rocío Fuente Pérez
- Universidad Europea de Madrid, Department of Nursing, Faculty of Medicine, Health and Sports, 28670 Madrid, Spain
| |
Collapse
|
8
|
Modafferi C, Tabolacci E, Lo Vecchio F, Cassano I, Bertozzi R, Fargnoli A, Cafiero C, Lo Cascio E, Arcovito A, Grippaudo C, Chiurazzi P. New Insight into the genotype-phenotype correlation of PTH1R variants and primary failure of tooth eruption on an Italian Cohort. Eur J Hum Genet 2024; 32:1402-1411. [PMID: 39327493 DOI: 10.1038/s41431-024-01691-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/01/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Primary failure of tooth eruption (PFE) is an autosomal dominant disease with penetrance defect. While the clinical phenotype is relatively well-defined since the 70 s of the last centuries, much more need to be clarified about the genetic causes of this condition. In our previous paper we established clinical criteria to better identify PFE patients carrying PTH1R gene variants. We examined a new cohort of 32 patients, including one or more relatives for 7 patients (43 cases in total), referred to have PFE and recruited from our Hospital and from external outpatients. Sequencing analysis of the PTH1R coding sequence in this cohort of patients revealed 9 different variants, 4 exonic and 5 intronic. Through in silico prediction tools and databases, 3 of them (2 exonic and 1 in a splicing site) had been considered potentially involved in the PFE phenotype. Sequencing of cDNA was unsuccessfully attempted due to the low levels of PTH1R expression in the analysed tissues. The yield of the genetic test increases when the clinical selection of the patients with dental eruption failure is well-characterized. Dental eruption failure with pure clinical findings of PFE associated with familial history revealed variants in PTH1R gene, offering a diagnostic test for the family. Characterization of novel variants in the most relevant responsible gene of the PFE could bring to a more accurate diagnosis and therapeutic approach in the future and to a deeper comprehension of the disease.
Collapse
Affiliation(s)
- Clarissa Modafferi
- UOC Genetica Medica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Elisabetta Tabolacci
- Dipartimento di Scienze della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Filomena Lo Vecchio
- UOC Genetica Medica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Ilaria Cassano
- UOC Genetica Medica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Roberto Bertozzi
- UOC Genetica Medica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Arcangelo Fargnoli
- UOC Genetica Medica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Concetta Cafiero
- Medical Oncology, SG Moscati Hospital, Statte, Ta, Italy
- Anatomic Pathology Unit, Fabrizio Spaziani Hospital, Frosinone, Italy
| | - Ettore Lo Cascio
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Sezione di Biochimica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Arcovito
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Sezione di Biochimica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Cristina Grippaudo
- UOC di Clinica Odontoiatrica, Dipartimento di Neuroscienze, Organi di senso e Torace, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy.
- Dipartimento Universitario Testa Collo ed Organi di Senso, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Pietro Chiurazzi
- UOC Genetica Medica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
- Dipartimento di Scienze della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| |
Collapse
|
9
|
Li XX, Wang MT, Wu ZF, Sun Q, Ono N, Nagata M, Zang XL, Ono W. Etiological Mechanisms and Genetic/Biological Modulation Related to PTH1R in Primary Failure of Tooth Eruption. Calcif Tissue Int 2024; 115:101-116. [PMID: 38833001 DOI: 10.1007/s00223-024-01227-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 05/07/2024] [Indexed: 06/06/2024]
Abstract
Primary failure of eruption (PFE) is a rare disorder that is characterized by the inability of a molar tooth/teeth to erupt to the occlusal plane or to normally react to orthodontic force. This condition is related to hereditary factors and has been extensively researched over many years. However, the etiological mechanisms of pathogenesis are still not fully understood. Evidence from studies on PFE cases has shown that PFE patients may carry parathyroid hormone 1 receptor (PTH1R) gene mutations, and genetic detection can be used to diagnose PFE at an early stage. PTH1R variants can lead to altered protein structure, impaired protein function, and abnormal biological activities of the cells, which may ultimately impact the behavior of teeth, as observed in PFE. Dental follicle cells play a critical role in tooth eruption and root development and are regulated by parathyroid hormone-related peptide (PTHrP)-PTH1R signaling in their differentiation and other activities. PTHrP-PTH1R signaling also regulates the activity of osteoblasts, osteoclasts and odontoclasts during tooth development and eruption. When interference occurs in the PTHrP-PTH1R signaling pathway, the normal function of dental follicles and bone remodeling are impaired. This review provides an overview of PTH1R variants and their correlation with PFE, and highlights that a disruption of PTHrP-PTH1R signaling impairs the normal process of tooth development and eruption, thus providing insight into the underlying mechanisms related to PTH1R and its role in driving PFE.
Collapse
Affiliation(s)
- Xiao-Xia Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Man-Ting Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Zhi-Fang Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Qiang Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Noriaki Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, 77054, USA
| | - Mizuki Nagata
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, 77054, USA
| | - Xiao-Long Zang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China.
| | - Wanida Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, 77054, USA.
| |
Collapse
|
10
|
Jacobsen C, Jüppner H, Mitchell DM. Case 21-2024: A 10-Month-Old Boy with Vomiting and Hypercalcemia. N Engl J Med 2024; 391:167-176. [PMID: 38986061 PMCID: PMC12044401 DOI: 10.1056/nejmcpc2402485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Affiliation(s)
- Christina Jacobsen
- From the Department of Pediatrics, Boston Children's Hospital (C.J.), the Department of Pediatrics, Massachusetts General Hospital (D.M.M., H.J.), and the Department of Pediatrics, Harvard Medical School (C.J., D.M.M., H.J.) - all in Boston
| | - Harald Jüppner
- From the Department of Pediatrics, Boston Children's Hospital (C.J.), the Department of Pediatrics, Massachusetts General Hospital (D.M.M., H.J.), and the Department of Pediatrics, Harvard Medical School (C.J., D.M.M., H.J.) - all in Boston
| | - Deborah M Mitchell
- From the Department of Pediatrics, Boston Children's Hospital (C.J.), the Department of Pediatrics, Massachusetts General Hospital (D.M.M., H.J.), and the Department of Pediatrics, Harvard Medical School (C.J., D.M.M., H.J.) - all in Boston
| |
Collapse
|
11
|
Walker V. The Intricacies of Renal Phosphate Reabsorption-An Overview. Int J Mol Sci 2024; 25:4684. [PMID: 38731904 PMCID: PMC11083860 DOI: 10.3390/ijms25094684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
To maintain an optimal body content of phosphorus throughout postnatal life, variable phosphate absorption from food must be finely matched with urinary excretion. This amazing feat is accomplished through synchronised phosphate transport by myriads of ciliated cells lining the renal proximal tubules. These respond in real time to changes in phosphate and composition of the renal filtrate and to hormonal instructions. How they do this has stimulated decades of research. New analytical techniques, coupled with incredible advances in computer technology, have opened new avenues for investigation at a sub-cellular level. There has been a surge of research into different aspects of the process. These have verified long-held beliefs and are also dramatically extending our vision of the intense, integrated, intracellular activity which mediates phosphate absorption. Already, some have indicated new approaches for pharmacological intervention to regulate phosphate in common conditions, including chronic renal failure and osteoporosis, as well as rare inherited biochemical disorders. It is a rapidly evolving field. The aim here is to provide an overview of our current knowledge, to show where it is leading, and where there are uncertainties. Hopefully, this will raise questions and stimulate new ideas for further research.
Collapse
Affiliation(s)
- Valerie Walker
- Department of Clinical Biochemistry, University Hospital Southampton NHS Foundation Trust, Southampton General Hospital, Southampton S016 6YD, UK
| |
Collapse
|
12
|
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
|
13
|
Miller WE, O'Connor CM. CMV-encoded GPCRs in infection, disease, and pathogenesis. Adv Virus Res 2024; 118:1-75. [PMID: 38461029 DOI: 10.1016/bs.aivir.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2024]
Abstract
G protein coupled receptors (GPCRs) are seven-transmembrane domain proteins that modulate cellular processes in response to external stimuli. These receptors represent the largest family of membrane proteins, and in mammals, their signaling regulates important physiological functions, such as vision, taste, and olfaction. Many organisms, including yeast, slime molds, and viruses encode GPCRs. Cytomegaloviruses (CMVs) are large, betaherpesviruses, that encode viral GPCRs (vGPCRs). Human CMV (HCMV) encodes four vGPCRs, including UL33, UL78, US27, and US28. Each of these vGPCRs, as well as their rodent and primate orthologues, have been investigated for their contributions to viral infection and disease. Herein, we discuss how the CMV vGPCRs function during lytic and latent infection, as well as our understanding of how they impact viral pathogenesis.
Collapse
Affiliation(s)
- William E Miller
- Department of Molecular and Cellular Bioscience, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Christine M O'Connor
- Infection Biology, Sheikha Fatima bint Mubarak Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States; Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland Clinic, Cleveland, OH, United States; Case Comprehensive Cancer Center, Cleveland, OH, United States.
| |
Collapse
|
14
|
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: 5] [Impact Index Per Article: 5.0] [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
|
15
|
Wei S, Pour NG, Tiruvadi-Krishnan S, Ray AP, Thakur N, Eddy MT, Lamichhane R. Single-molecule visualization of human A 2A adenosine receptor activation by a G protein and constitutively activating mutations. Commun Biol 2023; 6:1218. [PMID: 38036689 PMCID: PMC10689853 DOI: 10.1038/s42003-023-05603-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023] Open
Abstract
Mutations that constitutively activate G protein-coupled receptors (GPCRs), known as constitutively activating mutations (CAMs), modify cell signaling and interfere with drugs, resulting in diseases with limited treatment options. We utilize fluorescence imaging at the single-molecule level to visualize the dynamic process of CAM-mediated activation of the human A2A adenosine receptor (A2AAR) in real time. We observe an active-state population for all CAMs without agonist stimulation. Importantly, activating mutations significantly increase the population of an intermediate state crucial for receptor activation, notably distinct from the addition of a partner G protein. Activation kinetics show that while CAMs increase the frequency of transitions to the intermediate state, mutations altering sodium sensitivity increase transitions away from it. These findings indicate changes in GPCR function caused by mutations may be predicted based on whether they favor or disfavor formation of an intermediate state, providing a framework for designing receptors with altered functions or therapies that target intermediate states.
Collapse
Affiliation(s)
- Shushu Wei
- Department of Biochemistry & Cellular and Molecular Biology, College of Arts and Sciences, University of Tennessee, Knoxville, TN, USA
| | - Niloofar Gopal Pour
- Department of Chemistry, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL, USA
| | - Sriram Tiruvadi-Krishnan
- Department of Biochemistry & Cellular and Molecular Biology, College of Arts and Sciences, University of Tennessee, Knoxville, TN, USA
| | - Arka Prabha Ray
- Department of Chemistry, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL, USA
| | - Naveen Thakur
- Department of Chemistry, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL, USA
| | - Matthew T Eddy
- Department of Chemistry, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL, USA.
| | - Rajan Lamichhane
- Department of Biochemistry & Cellular and Molecular Biology, College of Arts and Sciences, University of Tennessee, Knoxville, TN, USA.
| |
Collapse
|
16
|
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: 0.5] [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
|
17
|
Amano K, Kitaoka Y, Kato S, Fujiwara M, Okuzaki D, Aikawa T, Kogo M, Iida S. Pth1r Signal in Gli1+ Cells Maintains Postnatal Cranial Base Synchondrosis. J Dent Res 2023; 102:1241-1251. [PMID: 37575041 DOI: 10.1177/00220345231184405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023] Open
Abstract
Cranial base synchondroses are the endochondral ossification centers for cranial base growth and thus indispensable for proper skull, brain, and midfacial development. The synchondroses are composed of mirror-image growth plates that are continuously maintained from the embryonic to postnatal stage through chondrocyte differentiation. Several factors, including Pth1r signaling, are known to control fetal synchondrosis development. However, there are currently no reports regarding any role for Pth1r signaling in postnatal cranial base and synchondrosis development. Also, the mesenchymal cells that source Pth1r signaling for synchondroses are not known. Here, we employed an inducible mouse model, a hedgehog-responsive Gli1-CreERT2 driver, focusing on the postnatal study. We performed 2 inducible protocols using Gli1-CreERT2;Tomatofl/+ mice that uncovered distinct patterning of Gli1-positive and Gli1-negative chondrocytes in the synchondrosis cartilage. Moreover, we generated Gli1-CreERT2;Pth1rfl/fl;Tomatofl/+ mice to assess their functions in postnatal synchondrosis and found that the mutants had survived postnatally. The mutant skulls morphologically presented unambiguous phenotypes where we noticed the shortened cranial base and premature synchondrosis closure. Histologically, gradual disorganization in mutant synchondroses caused an uncommon remaining central zone between hypertrophic zones on both sides while the successive differentiation of round, flat, and hypertrophic chondrocytes was observed in control sections. These mutant synchondroses disappeared and were finally replaced by bone. Of note, the mutant fusing synchondroses lost their characteristic patterning of Gli1-positive and Gli1-negative chondrocytes, suggesting that loss of Pth1r signaling alters the distribution of hedgehog-responsive chondrocytes. Moreover, we performed laser microdissection and RNA sequencing to characterize the flat proliferative and round resting chondrocytes where we found flat chondrocytes have a characteristic feature of both chondrocyte proliferation and maturation. Taken together, these data demonstrate that Pth1r signaling in Gli1-positive cells is essential for postnatal development and maintenance in cranial base synchondroses. Our findings will elucidate previously unknown aspects of Pth1r functions in cranial biology and development.
Collapse
Affiliation(s)
- K Amano
- Department of Oral and Maxillofacial Reconstructive Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- The first department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Y Kitaoka
- The first department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - S Kato
- Department of Oral and Maxillofacial Reconstructive Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - M Fujiwara
- The first department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Osaka, Japan
- The Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - D Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - T Aikawa
- The first department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - M Kogo
- The first department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - S Iida
- Department of Oral and Maxillofacial Reconstructive Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
18
|
Portales-Castillo I, Dean T, Cheloha RW, Creemer BA, Vilardaga JP, Savransky S, Khatri A, Jüppner H, Gardella TJ. Altered Signaling and Desensitization Responses in PTH1R Mutants Associated with Eiken Syndrome. Commun Biol 2023; 6:599. [PMID: 37268817 PMCID: PMC10238420 DOI: 10.1038/s42003-023-04966-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 05/22/2023] [Indexed: 06/04/2023] Open
Abstract
The parathyroid hormone receptor type 1 (PTH1R) is a G protein-coupled receptor that plays key roles in regulating calcium homeostasis and skeletal development via binding the ligands, PTH and PTH-related protein (PTHrP), respectively. Eiken syndrome is a rare disease of delayed bone mineralization caused by homozygous PTH1R mutations. Of the three mutations identified so far, R485X, truncates the PTH1R C-terminal tail, while E35K and Y134S alter residues in the receptor's amino-terminal extracellular domain. Here, using a variety of cell-based assays, we show that R485X increases the receptor's basal rate of cAMP signaling and decreases its capacity to recruit β-arrestin2 upon ligand stimulation. The E35K and Y134S mutations each weaken the binding of PTHrP leading to impaired β-arrestin2 recruitment and desensitization of cAMP signaling response to PTHrP but not PTH. Our findings support a critical role for interaction with β-arrestin in the mechanism by which the PTH1R regulates bone formation.
Collapse
Affiliation(s)
- Ignacio Portales-Castillo
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Thier Research Building, 50 Blossom St, Boston, MA, 02114, USA
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, and Harvard Medical School, Thier Research Building, 50 Blossom St, Boston, MA, 02114, USA
- Department of Medicine, Division of Nephrology, Washington University in St. Louis, BJCIH Building, 425 South Euclid St, St. Louis, MO, 63110, USA
| | - Thomas Dean
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Thier Research Building, 50 Blossom St, Boston, MA, 02114, USA
| | - Ross W Cheloha
- Chemical Biology in Signaling Section, Laboratory of Bioorganic Chemistry, National Institutes of Diabetes and Digestive and Kidney Diseases, Building 8, 8 Center Drive, Bethesda, MD, 20891, USA
| | - Brendan A Creemer
- Chemical Biology in Signaling Section, Laboratory of Bioorganic Chemistry, National Institutes of Diabetes and Digestive and Kidney Diseases, Building 8, 8 Center Drive, Bethesda, MD, 20891, USA
| | - Jean-Pierre Vilardaga
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Thomas E. Starzl Biomedical Science Tower, 200 Lothrop St, Pittsburgh, PA, 15261, USA
| | - Sofya Savransky
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Thomas E. Starzl Biomedical Science Tower, 200 Lothrop St, Pittsburgh, PA, 15261, USA
| | - Ashok Khatri
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Thier Research Building, 50 Blossom St, Boston, MA, 02114, USA
| | - Harald Jüppner
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Thier Research Building, 50 Blossom St, Boston, MA, 02114, USA
- Pediatric Nephrology Unit, Massachusetts General Hospital, and Harvard Medical School, Thier Research Building, 50 Blossom St, Boston, MA, 02114, USA
| | - Thomas J Gardella
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Thier Research Building, 50 Blossom St, Boston, MA, 02114, USA.
| |
Collapse
|
19
|
Zhang S, Li T, Feng Y, Zhang K, Zou J, Weng X, Yuan Y, Zhang L. Exercise improves subchondral bone microenvironment through regulating bone-cartilage crosstalk. Front Endocrinol (Lausanne) 2023; 14:1159393. [PMID: 37288291 PMCID: PMC10242115 DOI: 10.3389/fendo.2023.1159393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 05/04/2023] [Indexed: 06/09/2023] Open
Abstract
Articular cartilage degeneration has been proved to cause a variety of joint diseases, among which osteoarthritis is the most typical. Osteoarthritis is characterized by articular cartilage degeneration and persistent pain, which affects the quality of life of patients as well as brings a heavy burden to society. The occurrence and development of osteoarthritis is related to the disorder of the subchondral bone microenvironment. Appropriate exercise can improve the subchondral bone microenvironment, thus playing an essential role in preventing and treating osteoarthritis. However, the exact mechanism whereby exercise improves the subchondral bone microenvironment remains unclear. There is biomechanical interaction as well as biochemical crosstalk between bone and cartilage. And the crosstalk between bone and cartilage is the key to bone-cartilage homeostasis maintenance. From the perspective of biomechanical and biochemical crosstalk between bone and cartilage, this paper reviews the effects of exercise-mediated bone-cartilage crosstalk on the subchondral bone microenvironment, aiming to provide a theoretical basis for the prevention and treatment of degenerative bone diseases.
Collapse
Affiliation(s)
- Shihua Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- School of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Tingting Li
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Yao Feng
- School of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Keping Zhang
- School of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Jun Zou
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Xiquan Weng
- School of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Yu Yuan
- School of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Lan Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- College of Sports and Health, Shandong Sport University, Jinan, China
| |
Collapse
|
20
|
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: 14] [Impact Index Per Article: 7.0] [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
|
21
|
Brommage R, Liu J, Powell DR. Skeletal phenotypes in secreted frizzled-related protein 4 gene knockout mice mimic skeletal architectural abnormalities in subjects with Pyle's disease from SFRP4 mutations. Bone Res 2023; 11:9. [PMID: 36808149 PMCID: PMC9941579 DOI: 10.1038/s41413-022-00242-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/26/2022] [Accepted: 11/03/2022] [Indexed: 02/22/2023] Open
Abstract
Mutations in SFRP4 cause Pyle's bone disease with wide metaphyses and increased skeletal fragility. The WNT signaling pathway plays important roles in determining skeletal architecture and SFRP4 is a secreted Frizzled decoy receptor that inhibits WNT signaling. Seven cohorts of male and female Sfrp4 gene knockout mice, examined through 2 years of age, had a normal lifespan but showed cortical and trabecular bone phenotypes. Mimicking human Erlenmeyer flask deformities, bone cross-sectional areas were elevated 2-fold in the distal femur and proximal tibia but only 30% in femur and tibia shafts. Reduced cortical bone thickness was observed in the vertebral body, midshaft femur and distal tibia. Elevated trabecular bone mass and numbers were observed in the vertebral body, distal femur metaphysis and proximal tibia metaphysis. Midshaft femurs retained extensive trabecular bone through 2 years of age. Vertebral bodies had increased compressive strength, but femur shafts had reduced bending strength. Trabecular, but not cortical, bone parameters in heterozygous Sfrp4 mice were modestly affected. Ovariectomy resulted in similar declines in both cortical and trabecular bone mass in wild-type and Sfrp4 KO mice. SFRP4 is critical for metaphyseal bone modeling involved in determining bone width. Sfrp4 KO mice show similar skeletal architecture and bone fragility deficits observed in patients with Pyle's disease with SFRP4 mutations.
Collapse
Affiliation(s)
- Robert Brommage
- Department of Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, 77381, USA.
- BoneGenomics, The Woodlands, TX, USA.
| | - Jeff Liu
- Department of Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, 77381, USA.
- Biogen, Cambridge, MA, USA.
| | - David R Powell
- Department of Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, 77381, USA.
| |
Collapse
|
22
|
Csukasi F, Bosakova M, Barta T, Martin JH, Arcedo J, Barad M, Rico-Llanos GA, Zieba J, Becerra J, Krejci P, Duran I, Krakow D. Skeletal diseases caused by mutations in PTH1R show aberrant differentiation of skeletal progenitors due to dysregulation of DEPTOR. Front Cell Dev Biol 2023; 10:963389. [PMID: 36726589 PMCID: PMC9885499 DOI: 10.3389/fcell.2022.963389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 12/27/2022] [Indexed: 01/18/2023] Open
Abstract
Alterations in the balance between skeletogenesis and adipogenesis is a pathogenic feature in multiple skeletal disorders. Clinically, enhanced bone marrow adiposity in bones impairs mobility and increases fracture risk, reducing the quality of life of patients. The molecular mechanism that underlies the balance between skeletogenesis and adipogenesis is not completely understood but alterations in skeletal progenitor cells' differentiation pathway plays a key role. We recently demonstrated that parathyroid hormone (PTH)/PTH-related peptide (PTHrP) control the levels of DEPTOR, an inhibitor of the mechanistic target of rapamycin (mTOR), and that DEPTOR levels are altered in different skeletal diseases. Here, we show that mutations in the PTH receptor-1 (PTH1R) alter the differentiation of skeletal progenitors in two different skeletal genetic disorders and lead to accumulation of fat or cartilage in bones. Mechanistically, DEPTOR controls the subcellular localization of TAZ (transcriptional co-activator with a PDZ-binding domain), a transcriptional regulator that governs skeletal stem cells differentiation into either bone and fat. We show that DEPTOR regulation of TAZ localization is achieved through the control of Dishevelled2 (DVL2) phosphorylation. Depending on nutrient availability, DEPTOR directly interacts with PTH1R to regulate PTH/PTHrP signaling or it forms a complex with TAZ, to prevent its translocation to the nucleus and therefore inhibit its transcriptional activity. Our data point DEPTOR as a key molecule in skeletal progenitor differentiation; its dysregulation under pathologic conditions results in aberrant bone/fat balance.
Collapse
Affiliation(s)
- Fabiana Csukasi
- Department of Orthopaedic Surgery, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA, United States
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, University of Malaga, Institute of Biomedical Research in Malaga (IBIMA-Plataforma BIONAND), Malaga, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Michaela Bosakova
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia
- Institute of Animal Physiology and Genetics of the CAS, Brno, Czechia
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
| | - Tomas Barta
- Institute of Animal Physiology and Genetics of the CAS, Brno, Czechia
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Jorge H. Martin
- Department of Orthopaedic Surgery, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA, United States
| | - Jesus Arcedo
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, University of Malaga, Institute of Biomedical Research in Malaga (IBIMA-Plataforma BIONAND), Malaga, Spain
| | - Maya Barad
- Department of Orthopaedic Surgery, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA, United States
| | - Gustavo A. Rico-Llanos
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, University of Malaga, Institute of Biomedical Research in Malaga (IBIMA-Plataforma BIONAND), Malaga, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Jennifer Zieba
- Department of Orthopaedic Surgery, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA, United States
| | - Jose Becerra
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, University of Malaga, Institute of Biomedical Research in Malaga (IBIMA-Plataforma BIONAND), Malaga, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia
- Institute of Animal Physiology and Genetics of the CAS, Brno, Czechia
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
| | - Ivan Duran
- Department of Orthopaedic Surgery, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA, United States
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, University of Malaga, Institute of Biomedical Research in Malaga (IBIMA-Plataforma BIONAND), Malaga, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Deborah Krakow
- Department of Orthopaedic Surgery, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA, United States
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czechia
- Department of Human Genetics, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA, United States
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
23
|
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: 26] [Impact Index Per Article: 8.7] [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
|
24
|
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: 19] [Impact Index Per Article: 6.3] [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
|
25
|
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: 0.3] [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
|
26
|
Tiffany AS, Harley BAC. Growing Pains: The Need for Engineered Platforms to Study Growth Plate Biology. Adv Healthc Mater 2022; 11:e2200471. [PMID: 35905390 PMCID: PMC9547842 DOI: 10.1002/adhm.202200471] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/11/2022] [Indexed: 01/27/2023]
Abstract
Growth plates, or physis, are highly specialized cartilage tissues responsible for longitudinal bone growth in children and adolescents. Chondrocytes that reside in growth plates are organized into three distinct zones essential for proper function. Modeling key features of growth plates may provide an avenue to develop advanced tissue engineering strategies and perspectives for cartilage and bone regenerative medicine applications and a platform to study processes linked to disease progression. In this review, a brief introduction of the growth plates and their role in skeletal development is first provided. Injuries and diseases of the growth plates as well as physiological and pathological mechanisms associated with remodeling and disease progression are discussed. Growth plate biology, namely, its architecture and extracellular matrix organization, resident cell types, and growth factor signaling are then focused. Next, opportunities and challenges for developing 3D biomaterial models to study aspects of growth plate biology and disease in vitro are discussed. Finally, opportunities for increasingly sophisticated in vitro biomaterial models of the growth plate to study spatiotemporal aspects of growth plate remodeling, to investigate multicellular signaling underlying growth plate biology, and to develop platforms that address key roadblocks to in vivo musculoskeletal tissue engineering applications are described.
Collapse
Affiliation(s)
- Aleczandria S. Tiffany
- Department of Chemical and Biomolecular EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Brendan A. C. Harley
- Department of Chemical and Biomolecular EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
- Carl R. Woese Institute for Genomic BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| |
Collapse
|
27
|
Sarkar K, Joedicke L, Westwood M, Burnley R, Wright M, McMillan D, Byrne B. Modulation of PTH1R signaling by an extracellular binding antibody. VITAMINS AND HORMONES 2022; 120:109-132. [PMID: 35953107 DOI: 10.1016/bs.vh.2022.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Parathyroid hormone receptor 1 (PTH1R) is a class B G-protein coupled receptor with key roles in bone development. The receptor signals through both the Gs and Gq G-proteins as well as through β-arrestin in a G-protein independent manner. Current treatments for bone disorders, such as osteoporosis, target the PTH1R but are suboptimal in their efficacy. Monoclonal antibodies represent a major growth area in therapeutics as a result of their superior specificity and long serum half-life. Here, we discovered antibodies against the extracellular domain (ECD) of PTH1R from a phage display library. One of these antibodies, ECD-ScFvhFc, binds PTH1R with high affinity and although it has little or no effect on G-protein dependent receptor signaling, it does reduce PTH1R mediated β-arrestin signaling. Hydrogen-deuterium exchange mass spectrometry (HDX-MS) demonstrated that the ECD-ScFvhFc binding site overlapped partially with that of the cognate ligand, PTH. The results of this study demonstrate the suitability of PTH1R as a target for therapeutic antibody development.
Collapse
Affiliation(s)
- Kaushik Sarkar
- Department of Life Sciences, Imperial College, London, United Kingdom; UCB Pharma, Slough, United Kingdom
| | | | | | | | | | | | - Bernadette Byrne
- Department of Life Sciences, Imperial College, London, United Kingdom.
| |
Collapse
|
28
|
Yoon SH, Tang CC, Wein MN. Salt inducible kinases and PTH1R action. VITAMINS AND HORMONES 2022; 120:23-45. [PMID: 35953111 DOI: 10.1016/bs.vh.2022.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Parathyroid hormone is a central regulator of calcium homeostasis. PTH protects the organism from hypocalcemia through its actions in bone and kidney. Recent physiologic studies have revealed key target genes for PTH receptor (PTH1R) signaling in these target organs. However, the complete signal transduction cascade used by PTH1R to accomplish these physiologic actions has remained poorly defined. Here we will review recent studies that have defined an important role for salt inducible kinases downstream of PTH1R in bone, cartilage, and kidney. PTH1R signaling inhibits the activity of salt inducible kinases. Therefore, direct SIK inhibitors represent a promising novel strategy to mimic PTH actions using small molecules. Moreover, a detailed understanding of the molecular circuitry used by PTH1R to exert its biologic effects will afford powerful new models to better understand the diverse actions of this important G protein coupled receptor in health and disease.
Collapse
Affiliation(s)
- Sung-Hee Yoon
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Cheng-Chia Tang
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Marc N Wein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
29
|
Kimura T, Panaroni C, Rankin EB, Purton LE, Wu JY. Loss of Parathyroid Hormone Receptor Signaling in Osteoprogenitors Is Associated With Accumulation of Multiple Hematopoietic Lineages in the Bone Marrow. J Bone Miner Res 2022; 37:1321-1334. [PMID: 35490308 PMCID: PMC11479576 DOI: 10.1002/jbmr.4568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 04/20/2022] [Accepted: 04/26/2022] [Indexed: 11/10/2022]
Abstract
Osteoblasts and their progenitors play an important role in the support of hematopoiesis within the bone marrow (BM) microenvironment. We have previously reported that parathyroid hormone receptor (PTH1R) signaling in osteoprogenitors is required for normal B cell precursor differentiation, and for trafficking of maturing B cells out of the BM. Cells of the osteoblast lineage have been implicated in the regulation of several other hematopoietic cell populations, but the effects of PTH1R signaling in osteoprogenitors on other maturing hematopoietic populations have not been investigated. Here we report that numbers of maturing myeloid, T cell, and erythroid populations were increased in the BM of mice lacking PTH1R in Osx-expressing osteoprogenitors (PTH1R-OsxKO mice; knockout [KO]). This increase in maturing hematopoietic populations was not associated with an increase in progenitor populations or proliferation. The spleens of PTH1R-OsxKO mice were small with decreased numbers of all hematopoietic populations, suggesting that trafficking of mature hematopoietic populations between BM and spleen is impaired in the absence of PTH1R in osteoprogenitors. RNA sequencing (RNAseq) of osteoprogenitors and their descendants in bone and BM revealed increased expression of vascular cell adhesion protein 1 (VCAM-1) and C-X-C motif chemokine ligand 12 (CXCL12), factors that are involved in trafficking of several hematopoietic populations. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Takaharu Kimura
- Department of Medicine (Endocrinology), Stanford University School of Medicine, Stanford, CA, USA
| | - Cristina Panaroni
- Department of Medicine (Endocrinology), Stanford University School of Medicine, Stanford, CA, USA
| | - Erinn B Rankin
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Louise E Purton
- St Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
- The University of Melbourne, Department of Medicine at St Vincent's Hospital, Fitzroy, VIC, Australia
| | - Joy Y Wu
- Department of Medicine (Endocrinology), Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
30
|
Pathogenesis of FGF23-Related Hypophosphatemic Diseases Including X-linked Hypophosphatemia. ENDOCRINES 2022. [DOI: 10.3390/endocrines3020025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Since phosphate is indispensable for skeletal mineralization, chronic hypophosphatemia causes rickets and osteomalacia. Fibroblast growth factor 23 (FGF23), which is mainly produced by osteocytes in bone, functions as the central regulator of phosphate metabolism by increasing the renal excretion of phosphate and suppressing the production of 1,25-dihydroxyvitamin D. The excessive action of FGF23 results in hypophosphatemic diseases, which include a number of genetic disorders such as X-linked hypophosphatemic rickets (XLH) and tumor-induced osteomalacia (TIO). Phosphate-regulating gene homologous to endopeptidase on the X chromosome (PHEX), dentin matrix protein 1 (DMP1), ectonucleotide pyrophosphatase phosphodiesterase-1, and family with sequence similarity 20c, the inactivating variants of which are responsible for FGF23-related hereditary rickets/osteomalacia, are highly expressed in osteocytes, similar to FGF23, suggesting that they are local negative regulators of FGF23. Autosomal dominant hypophosphatemic rickets (ADHR) is caused by cleavage-resistant variants of FGF23, and iron deficiency increases serum levels of FGF23 and the manifestation of symptoms in ADHR. Enhanced FGF receptor (FGFR) signaling in osteocytes is suggested to be involved in the overproduction of FGF23 in XLH and autosomal recessive hypophosphatemic rickets type 1, which are caused by the inactivation of PHEX and DMP1, respectively. TIO is caused by the overproduction of FGF23 by phosphaturic tumors, which are often positive for FGFR. FGF23-related hypophosphatemia may also be associated with McCune-Albright syndrome, linear sebaceous nevus syndrome, and the intravenous administration of iron. This review summarizes current knowledge on the pathogenesis of FGF23-related hypophosphatemic diseases.
Collapse
|
31
|
Kurosaka H, Itoh S, Morita C, Tsujimoto T, Murata Y, Inubushi T, Yamashiro T. Development of dentition: From initiation to occlusion and related diseases. J Oral Biosci 2022; 64:159-164. [DOI: 10.1016/j.job.2022.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 12/19/2022]
|
32
|
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: 3.0] [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
|
33
|
Grippaudo C, D'Apolito I, Cafiero C, Re A, Chiurazzi P, Frazier-Bowers SA. Validating clinical characteristic of primary failure of eruption (PFE) associated with PTH1R variants. Prog Orthod 2021; 22:43. [PMID: 34897565 PMCID: PMC8666410 DOI: 10.1186/s40510-021-00387-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 10/23/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Primary failure of eruption (PFE) is a hereditary condition, and linkage with variants in the PTH1R gene has been demonstrated in many cases. The clinical severity and expression of PFE is variable, and the genotype-phenotype correlation remains elusive. Further, the similarity between some eruption disorders that are not associated with PTH1R alterations is striking. To better understand the genotype-phenotype correlation, we examined the relationship between the eruption phenotype and PTH1R genotype in 44 patients with suspected PFE and 27 unaffected relatives. Sanger sequencing was employed to analyze carefully selected PFE patients. Potential pathogenicity of variants was evaluated against multiple genetic databases for function prediction and frequency information. RESULTS Mutational analysis of the PTH1R coding sequence revealed 14 different variants in 38 individuals (30 patients and 8 first-degree relatives), 9 exonic and 5 intronic. Their pathogenicity has been reported and compared with the number and severity of clinical signs. In 72.7% of patients with pathogenic variants, five clinical and radiographic criteria have been found: involvement of posterior teeth, involvement of the distal teeth to the most mesial affected, supracrestal presentation, altered vertical growth of the alveolar process and posterior open-bite. In cases with mixed dentition (3), the deciduous molars of the affected quadrant were infraoccluded. DISCUSSION The probability of an affected patient having a PTH1R variant is greater when five specific clinical characteristics are present. The likelihood of an eruption defect in the absence of specific clinical characteristics is rarely associated with a PTH1R mutation. CONCLUSIONS We report here that systematic clinical and radiographic observation using a diagnostic rubric is highly valuable in confirming PFE and offers a reliable alternative for accurate diagnosis.
Collapse
Affiliation(s)
- Cristina Grippaudo
- School of Dentistry, Università Cattolica del Sacro Cuore, L.go Agostino Gemelli 8, 00168, Rome, Italy.,Fondazione Policlinico Universitario "A. Gemelli" IRCCS, L.go Agostino Gemelli 8, 00168, Rome, Italy
| | - Isabella D'Apolito
- School of Dentistry, Università Cattolica del Sacro Cuore, L.go Agostino Gemelli 8, 00168, Rome, Italy
| | | | - Agnese Re
- Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Pietro Chiurazzi
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy. .,UOC Genetica Medica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Largo Agostino Gemelli 8, 00168, Rome, Italy.
| | | |
Collapse
|
34
|
Motlaghzadeh Y, Bilezikian JP, Sellmeyer DE. Rare Causes of Hypercalcemia: 2021 Update. J Clin Endocrinol Metab 2021; 106:3113-3128. [PMID: 34240162 DOI: 10.1210/clinem/dgab504] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Primary hyperparathyroidism and malignancy are the etiologies in 90% of cases of hypercalcemia. When these entities are not the etiology of hypercalcemia, uncommon conditions need to be considered. In 2005, Jacobs and Bilezikian published a clinical review of rare causes of hypercalcemia, focusing on mechanisms and pathophysiology. This review is an updated synopsis of rare causes of hypercalcemia, extending the observations of the original article. EVIDENCE ACQUISITION Articles reporting rare associations between hypercalcemia and unusual conditions were identified through a comprehensive extensive PubMed-based search using the search terms "hypercalcemia" and "etiology," as well as examining the references in the identified case reports. We categorized the reports by adults vs pediatric and further categorized the adult reports based on etiology. Some included reports lacked definitive assessment of etiology and are reported as unknown mechanism with discussion of likely etiology. EVIDENCE SYNTHESIS There is a growing understanding of the breadth of unusual causes of hypercalcemia. When the cause of hypercalcemia is elusive, a focus on mechanism and review of prior reported cases is key to successful determination of the etiology. CONCLUSIONS The ever-expanding reports of patients with rare and even unknown mechanisms of hypercalcemia illustrate the need for continued investigation into the complexities of human calcium metabolism.
Collapse
Affiliation(s)
- Yasaman Motlaghzadeh
- Stanford University School of Medicine, Division of Endocrinology, Gerontology and Metabolism, Palo Alto, CA 94305, USA
| | - John P Bilezikian
- Division of Endocrinology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Deborah E Sellmeyer
- Stanford University School of Medicine, Division of Endocrinology, Gerontology and Metabolism, Palo Alto, CA 94305, USA
| |
Collapse
|
35
|
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: 65] [Impact Index Per Article: 16.3] [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
|
36
|
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: 1.5] [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
|
37
|
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
|
38
|
Schöneberg T, Liebscher I. Mutations in G Protein-Coupled Receptors: Mechanisms, Pathophysiology and Potential Therapeutic Approaches. Pharmacol Rev 2021; 73:89-119. [PMID: 33219147 DOI: 10.1124/pharmrev.120.000011] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
There are approximately 800 annotated G protein-coupled receptor (GPCR) genes, making these membrane receptors members of the most abundant gene family in the human genome. Besides being involved in manifold physiologic functions and serving as important pharmacotherapeutic targets, mutations in 55 GPCR genes cause about 66 inherited monogenic diseases in humans. Alterations of nine GPCR genes are causatively involved in inherited digenic diseases. In addition to classic gain- and loss-of-function variants, other aspects, such as biased signaling, trans-signaling, ectopic expression, allele variants of GPCRs, pseudogenes, gene fusion, and gene dosage, contribute to the repertoire of GPCR dysfunctions. However, the spectrum of alterations and GPCR involvement is probably much larger because an additional 91 GPCR genes contain homozygous or hemizygous loss-of-function mutations in human individuals with currently unidentified phenotypes. This review highlights the complexity of genomic alteration of GPCR genes as well as their functional consequences and discusses derived therapeutic approaches. SIGNIFICANCE STATEMENT: With the advent of new transgenic and sequencing technologies, the number of monogenic diseases related to G protein-coupled receptor (GPCR) mutants has significantly increased, and our understanding of the functional impact of certain kinds of mutations has substantially improved. Besides the classical gain- and loss-of-function alterations, additional aspects, such as biased signaling, trans-signaling, ectopic expression, allele variants of GPCRs, uniparental disomy, pseudogenes, gene fusion, and gene dosage, need to be elaborated in light of GPCR dysfunctions and possible therapeutic strategies.
Collapse
Affiliation(s)
- Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, Leipzig, Germany
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, Leipzig, Germany
| |
Collapse
|
39
|
Xiao X, Qin M, Zhang F, Su Y, Zhou B, Zhou Z. Understanding the Mechanism of Activation/Deactivation of GLP-1R via Accelerated Molecular Dynamics Simulation. Aust J Chem 2021. [DOI: 10.1071/ch20127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Glucagon-like peptide-1 receptor (GLP-1R), as a member of the class B G protein-coupled receptors (GPCRs), plays a crucial role in regulating blood glucose level signal recognition through its activation. The conformation changes during the activation pathway are of particular importance for its function. To investigate the activation mechanism of GLP-1R, the crystal structures of active and inactive forms are chosen to perform a total of 2 μs of accelerated molecular dynamics (aMD) simulations and 400ns of conventional molecular dynamics (cMD) simulations. With the aid of structural analysis and potential of mean force (PMF) calculations, we reveal the role of different helices in the activation and deactivation process and obtain the intermediate states during activation and deactivation that are difficult to capture in experiments. Protein structure network (PSN) was utilised to clarify the allosteric communication pathways of activation and deactivation and reveal the mechanisms of its activation and deactivation. The results could advance our understanding of the activation mechanism of GLP-1R and the related drug design.
Collapse
|
40
|
Feleke M, Bennett S, Chen J, Hu X, Williams D, Xu J. New physiological insights into the phenomena of deer antler: A unique model for skeletal tissue regeneration. J Orthop Translat 2020; 27:57-66. [PMID: 33437638 PMCID: PMC7773678 DOI: 10.1016/j.jot.2020.10.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/23/2020] [Accepted: 10/29/2020] [Indexed: 12/14/2022] Open
Abstract
Generally, mammals are unable to regenerate complex tissues and organs however the deer antler provides a rare anomaly to this rule. This osseous cranial appendage which is located on the frontal bone of male deer is capable of stem cell-based organogenesis, annual casting, and cyclic de novo regeneration. A series of recent studies have classified this form of regeneration as epimorphic stem cell based. Antler renewal is initiated by the activation of neural crest derived pedicle periosteal cells (PPCs) found residing within the pedicle periosteum (PP), these PPCs have the potential to differentiate into multiple lineages. Other antler stem cells (ASCs) are the reserve mesenchymal cells (RMCs) located in the antlers tip, which develop into cartilage tissue. Antlerogenic periosteal cells (APCs) found within the antlerogenic periosteum (AP) form the tissues of both the pedicle and first set of antlers. Antler stem cells (ASCs) further appear to progress through various stages of activation, this coordinated transition is considered imperative for stem cell-based mammalian regeneration. The latest developments have shown that the rapid elongation of the main beam and antler branches are a controlled form of tumour growth, regulated by the tumour suppressing genes TP73 and ADAMTS18. Both osteoclastogenesis, as well as osteogenic and chondrogenic differentiation are also involved. While there remains much to uncover this review both summarises and comprehensively evaluates our existing knowledge of tissue regeneration in the deer antler. This will assist in achieving the goal of in vitro organ regeneration in humans by furthering the field of modern regenerative medicine. The Translational potential of this article As a unique stem cell-based organ regeneration process in mammals, the deer antler represents a prime model system for investigating mechanisms of regeneration in mammalian tissues. Novel ASCs could provide cell-based therapies for regenerative medicine and bone remodelling for clinical application. A greater understanding of this process and a more in-depth defining of ASCs will potentiate improved clinical outcomes.
Collapse
Affiliation(s)
- Mesalie Feleke
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, 6009, Australia
| | - Samuel Bennett
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, 6009, Australia
| | - Jiazhi Chen
- Guangdong Provincial Key Laboratory of Industrial Surfactant, Guangdong Research Institute of Petrochemical and Fine Chemical Engineering, Guangdong Academy of Sciences, Guangzhou, 510665, China.,Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, 6009, Australia
| | - Xiaoyong Hu
- Guangdong Provincial Key Laboratory of Industrial Surfactant, Guangdong Research Institute of Petrochemical and Fine Chemical Engineering, Guangdong Academy of Sciences, Guangzhou, 510665, China
| | - Desmond Williams
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, 6009, Australia
| | - Jiake Xu
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, 6009, Australia
| |
Collapse
|
41
|
Serna J, Bergwitz C. Importance of Dietary Phosphorus for Bone Metabolism and Healthy Aging. Nutrients 2020; 12:E3001. [PMID: 33007883 PMCID: PMC7599912 DOI: 10.3390/nu12103001] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 12/13/2022] Open
Abstract
Inorganic phosphate (Pi) plays a critical function in many tissues of the body: for example, as part of the hydroxyapatite in the skeleton and as a substrate for ATP synthesis. Pi is the main source of dietary phosphorus. Reduced bioavailability of Pi or excessive losses in the urine causes rickets and osteomalacia. While critical for health in normal amounts, dietary phosphorus is plentiful in the Western diet and is often added to foods as a preservative. This abundance of phosphorus may reduce longevity due to metabolic changes and tissue calcifications. In this review, we examine how dietary phosphorus is absorbed in the gut, current knowledge about Pi sensing, and endocrine regulation of Pi levels. Moreover, we also examine the roles of Pi in different tissues, the consequences of low and high dietary phosphorus in these tissues, and the implications for healthy aging.
Collapse
Affiliation(s)
- Juan Serna
- Yale College, Yale University, New Haven, CT 06511, USA;
| | - Clemens Bergwitz
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06519, USA
| |
Collapse
|
42
|
Cryo-EM structure of an activated VIP1 receptor-G protein complex revealed by a NanoBiT tethering strategy. Nat Commun 2020; 11:4121. [PMID: 32807782 PMCID: PMC7431577 DOI: 10.1038/s41467-020-17933-8] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 07/17/2020] [Indexed: 02/06/2023] Open
Abstract
Vasoactive intestinal polypeptide receptor (VIP1R) is a widely expressed class B G protein-coupled receptor and a drug target for the treatment of neuronal, metabolic, and inflammatory diseases. However, our understanding of its mechanism of action and the potential of drug discovery targeting this receptor is limited by the lack of structural information of VIP1R. Here we report a cryo-electron microscopy structure of human VIP1R bound to PACAP27 and Gs heterotrimer, whose complex assembly is stabilized by a NanoBiT tethering strategy. Comparison with other class B GPCR structures reveals that PACAP27 engages VIP1R with its N-terminus inserting into the ligand binding pocket at the transmembrane bundle of the receptor, which subsequently couples to the G protein in a receptor-specific manner. This structure has provided insights into the molecular basis of PACAP27 binding and VIP receptor activation. The methodology of the NanoBiT tethering may help to provide structural information of unstable complexes. Vasoactive intestinal polypeptide receptor (VIP1R) is a widely expressed class B G protein-coupled receptor and a drug target for the treatment of inflammatory diseases. Here authors report a cryoelectron microscopy structure of human VIP1R bound to PACAP27 and Gs heterotrimer, which provides insights into PACAP27 binding and VIP receptor activation.
Collapse
|
43
|
Behrmann A, Zhong D, Li L, Cheng SL, Mead M, Ramachandran B, Sabaeifard P, Goodarzi M, Lemoff A, Kronenberg HM, Towler DA. PTH/PTHrP Receptor Signaling Restricts Arterial Fibrosis in Diabetic LDLR -/- Mice by Inhibiting Myocardin-Related Transcription Factor Relays. Circ Res 2020; 126:1363-1378. [PMID: 32160132 PMCID: PMC7524585 DOI: 10.1161/circresaha.119.316141] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 03/11/2020] [Indexed: 12/24/2022]
Abstract
RATIONALE The PTH1R (PTH [parathyroid hormone]/PTHrP [PTH-related protein] receptor) is expressed in vascular smooth muscle (VSM) and increased VSM PTH1R signaling mitigates diet-induced arteriosclerosis in LDLR-/- mice. OBJECTIVE To study the impact of VSM PTH1R deficiency, we generated mice SM22-Cre:PTH1R(fl/fl);LDLR-/- mice (PTH1R-VKO) and Cre-negative controls. METHODS AND RESULTS Immunofluorescence and Western blot confirmed PTH1R expression in arterial VSM that was reduced by Cre-mediated knockout. PTH1R-VKO cohorts exhibited increased aortic collagen accumulation in vivo, and VSM cultures from PTH1R-VKO mice elaborated more collagen (2.5-fold; P=0.01) with elevated Col3a1 and Col1a1 expression. To better understand these profibrotic responses, we performed mass spectrometry on nuclear proteins extracted from Cre-negative controls and PTH1R-VKO VSM. PTH1R deficiency reduced Gata6 but upregulated the MADS (MCM1, Agamous, Deficiens, and Srf DNA-binding domain)-box transcriptional co-regulator, Mkl-1 (megakaryoblastic leukemia [translocation] 1). Co-transfection assays (Col3a1 promoter-luciferase reporter) confirmed PTH1R-mediated inhibition and Mkl-1-mediated activation of Col3a1 transcription. Regulation mapped to a conserved hybrid CT(A/T)6GG MADS-box cognate in the Col3a1 promoter. Mutations of C/G in this motif markedly reduced Col3a1 transcriptional regulation by PTH1R and Mkl-1. Upregulation of Col3a1 and Col1a1 in PTH1R-VKO VSM was inhibited by small interfering RNA targeting Mkl1 and by treatment with the Mkl-1 antagonist CCG1423 or the Rock (Rho-associated coiled-coil containing protein kinase)-2 inhibitor KD025. Chromatin precipitation demonstrated that VSM PTH1R deficiency increased Mkl-1 binding to Col3a1 and Col1a1, but not TNF, promoters. Proteomic studies of plasma extracellular vesicles and VSM from PTH1R-VKO mice identified C1r (complement component 1, r) and C1s (complement component 1, s), complement proteins involved in vascular collagen metabolism, as potential biomarkers. VSM C1r protein and C1r message were increased with PTH1R deficiency, mediated by Mkl-1-dependent transcription and inhibited by CCG1423 or KD025. CONCLUSIONS PTH1R signaling restricts collagen production in the VSM lineage, in part, via Mkl-1 regulatory circuits that control collagen gene transcription. Strategies that maintain homeostatic VSM PTH1R signaling, as reflected in extracellular vesicle biomarkers of VSM PTH1R/Mkl-1 action, may help mitigate arteriosclerosis and vascular fibrosis.
Collapse
MESH Headings
- Animals
- Aorta/metabolism
- Aorta/pathology
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cells, Cultured
- Collagen Type I/genetics
- Collagen Type I/metabolism
- Collagen Type I, alpha 1 Chain
- Collagen Type III/genetics
- Collagen Type III/metabolism
- Diabetes Mellitus/genetics
- Diabetes Mellitus/metabolism
- Diabetes Mellitus/pathology
- Diet, High-Fat
- Disease Models, Animal
- Fibrosis
- Humans
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Parathyroid Hormone/metabolism
- Rats
- Receptor, Parathyroid Hormone, Type 1/deficiency
- Receptor, Parathyroid Hormone, Type 1/genetics
- Receptor, Parathyroid Hormone, Type 1/metabolism
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Signal Transduction
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription, Genetic
- Vascular Remodeling
Collapse
Affiliation(s)
- Abraham Behrmann
- Internal Medicine – Endocrine Division, UT Southwestern Medical Center, Dallas, TX 75390
| | - Dalian Zhong
- Internal Medicine – Endocrine Division, UT Southwestern Medical Center, Dallas, TX 75390
| | - Li Li
- Internal Medicine – Endocrine Division, UT Southwestern Medical Center, Dallas, TX 75390
| | - Su-Li Cheng
- Internal Medicine – Endocrine Division, UT Southwestern Medical Center, Dallas, TX 75390
| | - Megan Mead
- Internal Medicine – Endocrine Division, UT Southwestern Medical Center, Dallas, TX 75390
| | - Bindu Ramachandran
- Internal Medicine – Endocrine Division, UT Southwestern Medical Center, Dallas, TX 75390
| | - Parastoo Sabaeifard
- Internal Medicine – Endocrine Division, UT Southwestern Medical Center, Dallas, TX 75390
| | | | - Andrew Lemoff
- Biochemistry, UT Southwestern Medical Center, Dallas, TX 75390
| | - Henry M. Kronenberg
- Endocrine Unit, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114
| | - Dwight A. Towler
- Internal Medicine – Endocrine Division, UT Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
44
|
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: 8] [Impact Index Per Article: 1.6] [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
|
45
|
Gabbett MT, Jeavons CJ, Gray PH. Severe hypertension-An infantile feature of Jansen metaphyseal chondrodysplasia? Am J Med Genet A 2020; 182:768-772. [PMID: 31977144 DOI: 10.1002/ajmg.a.61494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/09/2020] [Accepted: 01/12/2020] [Indexed: 11/10/2022]
Abstract
Jansen metaphyseal chondrodysplasia (JMC) is a rare autosomal dominant skeletal dysplasia caused by gain-of-function mutations in the parathyroid hormone receptor 1 gene, PTH1R. We report on a patient presenting in the neonatal period with clinical signs of JMC in addition to severe hypertension. A pathogenic mutation in PTH1R was demonstrated, but investigations for hypertension yielded normal results. Hypertension has not been previously associated with JMC. Given aberration of the parathyroid hormone (PTH)/parathyroid-related protein pathway is the underlying pathogenic mechanism attributed to JMC, and also given evidence that hyperparathyroidism plays an important role in blood pressure homeostasis, we propose that hypertension is a hitherto unrecognized feature of JMC.
Collapse
Affiliation(s)
- Michael T Gabbett
- School of Biomedical Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia.,School of Medicine, Griffith University, Gold Coast, Queensland, Australia.,Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia.,Genetic Health Queensland, Metro North Hospital and Health Service, Brisbane, Queensland, Australia
| | - Cassandra J Jeavons
- Genetic Health Queensland, Metro North Hospital and Health Service, Brisbane, Queensland, Australia.,Department of Medical Imaging, Children's Health Queensland, Brisbane, Queensland, Australia
| | - Peter H Gray
- Newborn Services, Mater Health Services, Brisbane, Queensland, Australia.,Mater Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
46
|
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: 37] [Impact Index Per Article: 6.2] [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
|
47
|
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: 49] [Impact Index Per Article: 8.2] [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
|
48
|
Nagata M, Ono N, Ono W. Mesenchymal Progenitor Regulation of Tooth Eruption: A View from PTHrP. J Dent Res 2019; 99:133-142. [PMID: 31623502 DOI: 10.1177/0022034519882692] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Tooth eruption is a unique biological process by which highly mineralized tissues emerge into the outer world, and it occurs concomitantly with tooth root formation. These 2 processes have been considered independent phenomena; however, recent studies support the theory that they are indeed intertwined. Dental mesenchymal progenitor cells in the dental follicle lie at the heart of the coupling of these 2 processes, providing a source for diverse mesenchymal cells that support formation of the highly functional tooth root and the periodontal attachment apparatus, while facilitating formation of osteoclasts. These cells are regulated by autocrine signaling by parathyroid hormone-related protein (PTHrP) and its parathyroid hormone/PTHrP receptor PPR. This PTHrP-PPR signaling appears to crosstalk with other signaling pathways and regulates proper cell fates of mesenchymal progenitor cell populations. Disruption of this autocrine PTHrP-PPR signaling in these cells leads to defective formation of the periodontal attachment apparatus, tooth root malformation, and failure of tooth eruption in molars, which essentially recapitulate primary failure of eruption in humans, a rare genetic disorder exclusively affecting tooth eruption. Diversity and distinct functionality of these mesenchymal progenitor cell populations that regulate tooth eruption and tooth root formation are beginning to be unraveled.
Collapse
Affiliation(s)
- M Nagata
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - N Ono
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - W Ono
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
49
|
Csukasi F, Duran I, Barad M, Barta T, Gudernova I, Trantirek L, Martin JH, Kuo CY, Woods J, Lee H, Cohn DH, Krejci P, Krakow D. The PTH/PTHrP-SIK3 pathway affects skeletogenesis through altered mTOR signaling. Sci Transl Med 2019; 10:10/459/eaat9356. [PMID: 30232230 DOI: 10.1126/scitranslmed.aat9356] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 08/31/2018] [Indexed: 12/19/2022]
Abstract
Studies have suggested a role for the mammalian (or mechanistic) target of rapamycin (mTOR) in skeletal development and homeostasis, yet there is no evidence connecting mTOR with the key signaling pathways that regulate skeletogenesis. We identified a parathyroid hormone (PTH)/PTH-related peptide (PTHrP)-salt-inducible kinase 3 (SIK3)-mTOR signaling cascade essential for skeletogenesis. While investigating a new skeletal dysplasia caused by a homozygous mutation in the catalytic domain of SIK3, we observed decreased activity of mTOR complex 1 (mTORC1) and mTORC2 due to accumulation of DEPTOR, a negative regulator of both mTOR complexes. This SIK3 syndrome shared skeletal features with Jansen metaphyseal chondrodysplasia (JMC), a disorder caused by constitutive activation of the PTH/PTHrP receptor. JMC-derived chondrocytes showed reduced SIK3 activity, elevated DEPTOR, and decreased mTORC1 and mTORC2 activity, indicating a common mechanism of disease. The data demonstrate that SIK3 is an essential positive regulator of mTOR signaling that functions by triggering DEPTOR degradation in response to PTH/PTHrP signaling during skeletogenesis.
Collapse
Affiliation(s)
- Fabiana Csukasi
- Department of Orthopaedic Surgery, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Ivan Duran
- Department of Orthopaedic Surgery, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Maya Barad
- Department of Orthopaedic Surgery, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Tomas Barta
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Iva Gudernova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Lukas Trantirek
- Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic
| | - Jorge H Martin
- Department of Orthopaedic Surgery, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Caroline Y Kuo
- Department of Pediatrics, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Jeremy Woods
- Department of Pediatrics, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Hane Lee
- Department of Pathology and Laboratory Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Daniel H Cohn
- Department of Orthopaedic Surgery, University of California-Los Angeles, Los Angeles, CA 90095, USA.,Orthopaedic Institute for Children, University of California-Los Angeles, Los Angeles, CA 90095, USA.,Department of Molecular, Cell and Developmental Biology, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Pavel Krejci
- Department of Orthopaedic Surgery, University of California-Los Angeles, Los Angeles, CA 90095, USA.,Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, 65691 Brno, Czech Republic.,Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, 60200 Brno, Czech Republic
| | - Deborah Krakow
- Department of Orthopaedic Surgery, University of California-Los Angeles, Los Angeles, CA 90095, USA. .,Orthopaedic Institute for Children, University of California-Los Angeles, Los Angeles, CA 90095, USA.,Department of Human Genetics, University of California-Los Angeles, Los Angeles, CA 90095, USA.,Department of Obstetrics and Gynecology, University of California-Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
50
|
Modulation of PTH1R signaling by an ECD binding antibody results in inhibition of β-arrestin 2 coupling. Sci Rep 2019; 9:14432. [PMID: 31594997 PMCID: PMC6783463 DOI: 10.1038/s41598-019-51016-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/24/2019] [Indexed: 02/05/2023] Open
Abstract
Parathyroid hormone receptor 1 (PTH1R) belongs to the secretin class of G protein coupled receptors (GPCRs) and natively binds parathyroid hormone (PTH) and parathyroid hormone related peptide (PTHrP). Ligand binding to PTH1R involves binding to the large extracellular domain (ECD) and the orthosteric pocket, inducing conformational changes in the transmembrane domain and receptor activation. PTH1R regulates bone metabolism, signaling mainly through Gs and Gq/11 G-proteins. Here, we used phage display to generate PTH1R ECD-specific antibodies with the aim of modulating receptor functionality. We identified ECD-scFvhFc, which exhibited high affinity binding to both the isolated ECD and to the full-length receptor in styrene-maleic acid (SMA) lipid particles. Epitope mapping using hydrogen-deuterium exchange mass spectrometry (HDX-MS) indicates that the α1 helix of the ECD is ECD-scFvhFc’s epitope which may partially overlap with the known PTH (1–34) binding site. However, PTH (1–34)-mediated Gs activation is Undisturbed by ECD-scFvhFc binding. In contrast, ECD-scFvhFc potently inhibits β-arrestin-2 recruitment after PTH (1–34)-driven receptor activation and thus represents the first monoclonal antibody to selectively inhibit distinct PTH1R signaling pathways. Given the complexity of PTH1R signaling and the emerging importance of biased GPCR activation in drug development, ECD-scFvhFc could be a valuable tool to study PTH1R signaling bias.
Collapse
|