1
|
He Z, Zhu Z, Tang T, Wang F, Guo P, Li J, Tung NTC, Liang Q, Liu S, Gao M, Liu X, Zhou Z. Enpp1 mutations promote upregulation of hedgehog signaling in heterotopic ossification with aging. J Bone Miner Metab 2024; 42:681-698. [PMID: 39212714 PMCID: PMC11632054 DOI: 10.1007/s00774-024-01543-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Heterotopic ossification of the tendon and ligament (HOTL) is a chronic progressive disease that is usually accompanied by thickening and ossification of ligaments and high osteogenic activity of the surrounding ligament tissue. However, the molecular mechanism of maintaining the cellular phenotype of HOTL remains unclear. MATERIALS AND METHODS We first constructed a model of HOTL, Enpp1flox/flox/EIIa-Cre mice, a novel genetic mouse system. Imaging, histological, and cell-level analyses were performed to investigate the progressive ossification of the posterior longitudinal ligament, Achilles tendons, and degeneration joints caused by Enpp1 deficiency. RESULTS The results indicate that Enpp1 deficiency led to markedly progressive heterotopic ossification (HO), especially spine, and Achilles tendons, and was associated with progressive degeneration of the knees. The bone mass was decreased in the long bone. Furthermore, fibroblasts from Enpp1flox/flox/EIIa-Cre mice had greater osteogenic differentiation potential following induction by osteogenesis, accompanied by enhanced hedgehog (Hh) signaling. In addition, fibroblast cells show senescence, and aggravation of the senescence phenotype by further osteogenic induction. CONCLUSION Our study indicated that with increasing age, mutations in Enpp1 promote ectopic ossification of spinal ligaments and endochondral ossification in tendons and further aggravate knee degeneration by upregulating hedgehog signaling.
Collapse
Affiliation(s)
- Zhongyuan He
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 517108, China
- Department of Orthopaedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhengya Zhu
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 517108, China
| | - Tao Tang
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 517108, China
| | - Fuan Wang
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 517108, China
| | - Peng Guo
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 517108, China
| | - Jianfeng Li
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 517108, China
| | - Nguyen Tran Canh Tung
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Qian Liang
- Department of Spine Surgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518025, China
| | - Shaoyu Liu
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510062, China
| | - ManMan Gao
- Department of Sport Medicine, Inst Translat Med, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518025, China.
| | - Xizhe Liu
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510062, China.
| | - Zhiyu Zhou
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 517108, China.
| |
Collapse
|
2
|
Seefried L. Clinical presentation and burden of ENPP1 deficiency in adults. Arch Pediatr 2024; 31:4S33-4S36. [PMID: 39343472 DOI: 10.1016/s0929-693x(24)00155-6] [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] [Indexed: 10/01/2024]
Abstract
While the clinical consequences of severe ENPP1 deficiency leading to the rare disorders generalized arterial calcification of infancy (GACI) and autosomal recessive hypophosphatemic rickets type 2 (ARHR2) are well defined and understood, much less is known about how this evolves into adulthood and how moderate ENPP1 deficiency can first manifest in adulthood. Moreover, growing evidence substantiates an association of genetic variants in the ENPP1 gene with a wide range of further clinical manifestations including early-onset osteoporosis, osteoarthritis, and different forms of spinal ligament calcifications, i.e., diffuse idiopathic skeletal hyperostosis (DISH) and ossification of the posterior/anterior longitudinal ligament (OPLL/OALL). Furthermore, conditions with primarily extraskeletal signs and symptoms such as Cole disease, coagulopathies, and metabolic syndrome can seemingly result from ENPP1 variants. The causality and the pathophysiology behind these different clinical presentations appear complex and require further research, especially since the coincidence of these different phenotypes is rarely described and available evidence suggests that part of the aforementioned manifestations may result from ENPP1 effects beyond the catalytic activity of processing ATP to AMP and inorganic pyrophosphate (PPi). Growing awareness of the additional ENPP1-related manifestations across the lifespan will advance our understanding of this complex condition and help to standardize diagnostic approaches and develop individually tailored treatment concepts.
Collapse
Affiliation(s)
- Lothar Seefried
- Musculoskeletal Center Würzburg, University Hospital Würzburg, Brettreichstr. 11, 97074 Würzburg, Germany.
| |
Collapse
|
3
|
Du B, Ru J, Zhan Z, Lin C, Liu Y, Mao W, Zhang J. Insight into small-molecule inhibitors targeting extracellular nucleotide pyrophosphatase/phosphodiesterase1 for potential multiple human diseases. Eur J Med Chem 2024; 268:116286. [PMID: 38432057 DOI: 10.1016/j.ejmech.2024.116286] [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: 12/31/2023] [Revised: 02/06/2024] [Accepted: 02/24/2024] [Indexed: 03/05/2024]
Abstract
Extracellular nucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) has been identified as a type II transmembrane glycoprotein. It plays a crucial role in various biological processes, such as bone mineralization, cancer cell proliferation, and immune regulation. Consequently, ENPP1 has garnered attention as a promising target for pharmacological interventions. Despite its potential, the development of clinical-stage ENPP1 inhibitors for solid tumors, diabetes, and silent rickets remains limited. However, there are encouraging findings from preclinical trials involving small molecules exhibiting favorable therapeutic effects and safety profiles. This perspective aims to shed light on the structural properties, biological functions and the relationship between ENPP1 and diseases. Additionally, it focuses on the structure-activity relationship of ENPP1 inhibitors, with the intention of guiding the future development of new and effective ENPP1 inhibitors.
Collapse
Affiliation(s)
- Baochan Du
- Department of Neurology, Neuro-system and Multimorbidity Laboratory and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinxiao Ru
- Department of Neurology, Neuro-system and Multimorbidity Laboratory and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zixuan Zhan
- Department of Neurology, Neuro-system and Multimorbidity Laboratory and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Congcong Lin
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yang Liu
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Wuyu Mao
- Department of Neurology, Neuro-system and Multimorbidity Laboratory and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Jifa Zhang
- Department of Neurology, Neuro-system and Multimorbidity Laboratory and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
4
|
Tourkova IL, Larrouture QC, Liu S, Luo J, Schlesinger PH, Blair HC. The ecto-nucleotide pyrophosphatase/phosphodiesterase 2 promotes early osteoblast differentiation and mineralization in stromal stem cells. Am J Physiol Cell Physiol 2024; 326:C843-C849. [PMID: 38223929 PMCID: PMC11193479 DOI: 10.1152/ajpcell.00692.2023] [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: 12/14/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/16/2024]
Abstract
The phosphodiesterase enzymes mediate calcium-phosphate deposition in various tissues, although which enzymes are active in bone mineralization is unclear. Using gene array analysis, we found that a member of ecto-nucleotide pyrophosphatase/phosphodiesterase family, ENPP2, was strongly down-regulated with age in stromal stem cells that produce osteoblasts and make bone. This is in keeping with reduced bone formation in older animals. Thus, we hypothesized that ENPP2 is, at least in part, an early mediator of bone formation and thus may reflect reduced bone formation with age. Since ENPP2 has not previously been shown to have a role in osteoblast differentiation, we studied its effect on bone differentiation from stromal stem cells, verified by flow cytometry for stem cell antigens. In these remarkably uniform osteoblast precursors, we did transfection with ENPP2 DsiRNA, scrambled DsiRNA, or no transfection to make cells with normal or greatly reduced ENPP2 and analyzed osteoblast differentiation and mineralization. Osteoblast differentiation down-regulation was shown by alizarin red binding, silver staining, and alkaline phosphatase activity. Differences were confirmed by real-time PCR for alkaline phosphatase (ALPL), osteocalcin (BGLAP), and ENPP2 and by Western Blot for Enpp2. These were decreased, ∼50%, in osteoblasts transfected with ENPP2 DsiRNA compared with cells transfected with a scrambled DsiRNA or not transfected (control) cells. This finding is the first evidence for the role of ENPP2 in osteoblast differentiation and mineralization.NEW & NOTEWORTHY We report the discovery that the ecto-nucleotide pyrophosphatase/phosphodiesterase, ENPP2, is an important regulator of early differentiation of bone-forming osteoblasts.
Collapse
Affiliation(s)
- Irina L Tourkova
- Research Service, VA Medical Centre, Pittsburgh, Pennsylvania, United States
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Quitterie C Larrouture
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Silvia Liu
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Jianhua Luo
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Paul H Schlesinger
- Department of Cell Biology, Washington University, St. Louis, Missouri, United States
| | - Harry C Blair
- Research Service, VA Medical Centre, Pittsburgh, Pennsylvania, United States
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
5
|
Kauffenstein G, Martin L, Le Saux O. The Purinergic Nature of Pseudoxanthoma Elasticum. BIOLOGY 2024; 13:74. [PMID: 38392293 PMCID: PMC10886499 DOI: 10.3390/biology13020074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/13/2024] [Accepted: 01/19/2024] [Indexed: 02/24/2024]
Abstract
Pseudoxanthoma Elasticum (PXE) is an inherited disease characterized by elastic fiber calcification in the eyes, the skin and the cardiovascular system. PXE results from mutations in ABCC6 that encodes an ABC transporter primarily expressed in the liver and kidneys. It took nearly 15 years after identifying the gene to better understand the etiology of PXE. ABCC6 function facilitates the efflux of ATP, which is sequentially hydrolyzed by the ectonucleotidases ENPP1 and CD73 into pyrophosphate (PPi) and adenosine, both inhibitors of calcification. PXE, together with General Arterial Calcification of Infancy (GACI caused by ENPP1 mutations) as well as Calcification of Joints and Arteries (CALJA caused by NT5E/CD73 mutations), forms a disease continuum with overlapping phenotypes and shares steps of the same molecular pathway. The explanation of these phenotypes place ABCC6 as an upstream regulator of a purinergic pathway (ABCC6 → ENPP1 → CD73 → TNAP) that notably inhibits mineralization by maintaining a physiological Pi/PPi ratio in connective tissues. Based on a review of the literature and our recent experimental data, we suggest that PXE (and GACI/CALJA) be considered as an authentic "purinergic disease". In this article, we recapitulate the pathobiology of PXE and review molecular and physiological data showing that, beyond PPi deficiency and ectopic calcification, PXE is associated with wide and complex alterations of purinergic systems. Finally, we speculate on the future prospects regarding purinergic signaling and other aspects of this disease.
Collapse
Affiliation(s)
- Gilles Kauffenstein
- UMR INSERM 1260, Regenerative Nanomedicine, University of Strasbourg, 67084 Strasbourg, France
| | - Ludovic Martin
- PXE Consultation Center, MAGEC Nord Reference Center for Rare Skin Diseases, Angers University Hospital, 49000 Angers, France
- MITOVASC-UMR CNRS 6015 INSERM 1083, University of Angers, 49000 Angers, France
| | - Olivier Le Saux
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| |
Collapse
|
6
|
Ferreira CR, Carpenter TO, Braddock DT. ENPP1 in Blood and Bone: Skeletal and Soft Tissue Diseases Induced by ENPP1 Deficiency. ANNUAL REVIEW OF PATHOLOGY 2024; 19:507-540. [PMID: 37871131 PMCID: PMC11062289 DOI: 10.1146/annurev-pathmechdis-051222-121126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The enzyme ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) codes for a type 2 transmembrane glycoprotein that hydrolyzes extracellular ATP to generate pyrophosphate (PPi) and adenosine monophosphate, thereby contributing to downstream purinergic signaling pathways. The clinical phenotypes induced by ENPP1 deficiency are seemingly contradictory and include early-onset osteoporosis in middle-aged adults and life-threatening vascular calcifications in the large arteries of infants with generalized arterial calcification of infancy. The progressive overmineralization of soft tissue and concurrent undermineralization of skeleton also occur in the general medical population, where it is referred to as paradoxical mineralization to highlight the confusing pathophysiology. This review summarizes the clinical presentation and pathophysiology of paradoxical mineralization unveiled by ENPP1 deficiency and the bench-to-bedside development of a novel ENPP1 biologics designed to treat mineralization disorders in the rare disease and general medical population.
Collapse
Affiliation(s)
- Carlos R Ferreira
- Metabolic Medicine Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Thomas O Carpenter
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Demetrios T Braddock
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA;
| |
Collapse
|
7
|
Bourne LE, Davies BK, Millan JL, Arnett TR, Wheeler-Jones CPD, Keen JAC, Roberts SJ, Orriss IR. Evidence that pyrophosphate acts as an extracellular signalling molecule to exert direct functional effects in primary cultures of osteoblasts and osteoclasts. Bone 2023; 176:116868. [PMID: 37549801 DOI: 10.1016/j.bone.2023.116868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Extracellular pyrophosphate (PPi) is well known for its fundamental role as a physiochemical mineralisation inhibitor. However, information about its direct actions on bone cells remains limited. This study shows that PPi decreased osteoclast formation and resorptive activity by ≤50 %. These inhibitory actions were associated with reduced expression of genes involved in osteoclastogenesis (Tnfrsf11a, Dcstamp) and bone resorption (Ctsk, Car2, Acp5). In osteoblasts, PPi present for the entire (0-21 days) or latter stages of culture (7-21/14-21 days) decreased bone mineralisation by ≤95 %. However, PPi present for the differentiation phase only (0-7/0-14 days) increased bone formation (≤70 %). Prolonged treatment with PPi resulted in earlier matrix deposition and increased soluble collagen levels (≤2.3-fold). Expression of osteoblast (RUNX2, Bglap) and early osteocyte (E11, Dmp1) genes along with mineralisation inhibitors (Spp1, Mgp) was increased by PPi (≤3-fold). PPi levels are regulated by tissue non-specific alkaline phosphatase (TNAP) and ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (NPP1). PPi reduced NPP1 expression in both cell types whereas TNAP expression (≤2.5-fold) and activity (≤35 %) were increased in osteoblasts. Breakdown of extracellular ATP by NPP1 represents a key source of PPi. ATP release from osteoclasts and osteoblasts was decreased ≤60 % by PPi and by a selective TNAP inhibitor (CAS496014-12-2). Pertussis toxin, which prevents Gαi subunit activation, was used to investigate whether G-protein coupled receptor (GPCR) signalling mediates the effects of PPi. The actions of PPi on bone mineralisation, collagen production, ATP release, gene/protein expression and osteoclast formation were abolished or attenuated by pertussis toxin. Together these findings show that PPi, modulates differentiation, function and gene expression in osteoblasts and osteoclasts. The ability of PPi to alter ATP release and NPP1/TNAP expression and activity indicates that cells can detect PPi levels and respond accordingly. Our data also raise the possibility that some actions of PPi on bone cells could be mediated by a Gαi-linked GPCR.
Collapse
Affiliation(s)
- Lucie E Bourne
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK; School of Applied Sciences, University of Brighton, UK
| | - Bethan K Davies
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK; Clinical and Experimental Endocrinology, KU, Leuven, Belgium
| | - Jose Luis Millan
- Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, USA
| | - Timothy R Arnett
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK; Department of Cell and Developmental Biology, University College London, London, UK
| | | | - Jacob A C Keen
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Scott J Roberts
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK.
| |
Collapse
|
8
|
Lopez V, Schuh HJM, Mirza S, Vaaßen VJ, Schmidt MS, Sylvester K, Idris RM, Renn C, Schäkel L, Pelletier J, Sévigny J, Naggi A, Scheffler B, Lee SY, Bendas G, Müller CE. Heparins are potent inhibitors of ectonucleotide pyrophosphatase/phospho-diesterase-1 (NPP1) - a promising target for the immunotherapy of cancer. Front Immunol 2023; 14:1173634. [PMID: 37711611 PMCID: PMC10497752 DOI: 10.3389/fimmu.2023.1173634] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 07/03/2023] [Indexed: 09/16/2023] Open
Abstract
Introduction Heparins, naturally occurring glycosaminoglycans, are widely used for thrombosis prevention. Upon application as anticoagulants in cancer patients, heparins were found to possess additional antitumor activities. Ectonucleotidases have recently been proposed as novel targets for cancer immunotherapy. Methods and results In the present study, we discovered that heparin and its derivatives act as potent, selective, allosteric inhibitors of the poorly investigated ectonucleotidase NPP1 (nucleotide pyrophosphatase/phosphodiesterase-1, CD203a). Structure-activity relationships indicated that NPP1 inhibition could be separated from the compounds' antithrombotic effect. Moreover, unfractionated heparin (UFH) and different low molecular weight heparins (LMWHs) inhibited extracellular adenosine production by the NPP1-expressing glioma cell line U87 at therapeutically relevant concentrations. As a consequence, heparins inhibited the ability of U87 cell supernatants to induce CD4+ T cell differentiation into immunosuppressive Treg cells. Discussion NPP1 inhibition likely contributes to the anti-cancer effects of heparins, and their specific optimization may lead to improved therapeutics for the immunotherapy of cancer.
Collapse
Affiliation(s)
- Vittoria Lopez
- Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
- PharmaCenter Bonn, University of Bonn, Bonn, Germany
| | - H. J. Maximilian Schuh
- Pharmaceutical Institute, Pharmaceutical and Cell Biological Chemistry, University of Bonn, Bonn, Germany
| | - Salahuddin Mirza
- Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
- PharmaCenter Bonn, University of Bonn, Bonn, Germany
| | - Victoria J. Vaaßen
- Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
- PharmaCenter Bonn, University of Bonn, Bonn, Germany
| | - Michael S. Schmidt
- Pharmaceutical Institute, Pharmaceutical and Cell Biological Chemistry, University of Bonn, Bonn, Germany
| | - Katharina Sylvester
- Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
- PharmaCenter Bonn, University of Bonn, Bonn, Germany
| | - Riham M. Idris
- Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
- PharmaCenter Bonn, University of Bonn, Bonn, Germany
| | - Christian Renn
- Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
- PharmaCenter Bonn, University of Bonn, Bonn, Germany
| | - Laura Schäkel
- Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
- PharmaCenter Bonn, University of Bonn, Bonn, Germany
| | - Julie Pelletier
- Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Jean Sévigny
- Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Départment de Microbiologie-Infectiologie et d’Immunologie, Faculté de Médecine, Université Laval, Quebec, QC, Canada
| | - Annamaria Naggi
- Institute for Chemical and Biochemical Research “G. Ronzoni”, Milan, Italy
| | - Björn Scheffler
- DKFZ Division Translational Neurooncology at the West German Cancer Center (WTZ), DKTK Partner site, University Hospital Essen and German Cancer Research Center, Heidelberg, Germany
| | - Sang-Yong Lee
- Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
- PharmaCenter Bonn, University of Bonn, Bonn, Germany
| | - Gerd Bendas
- Pharmaceutical Institute, Pharmaceutical and Cell Biological Chemistry, University of Bonn, Bonn, Germany
| | - Christa E. Müller
- Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
- PharmaCenter Bonn, University of Bonn, Bonn, Germany
| |
Collapse
|
9
|
Disorders of phosphate homeostasis in children, part 1: primer on mineral ion homeostasis and the roles of phosphate in skeletal biology. Pediatr Radiol 2022; 52:2278-2289. [PMID: 35536415 DOI: 10.1007/s00247-022-05374-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 01/22/2022] [Accepted: 04/01/2022] [Indexed: 10/18/2022]
Abstract
Phosphate has extensive physiological roles including energy metabolism, genetic function, signal transduction and membrane integrity. Regarding the skeleton, not only do phosphate and calcium form the mineral component of the skeleton, but phosphate is also essential in regulating function of skeletal cells. Although our understanding of phosphate homeostasis has lagged behind and remains less than that for calcium, considerable advances have been made since the recognition of fibroblast growth factor-23 (FGF23) as a bone-derived phosphaturic hormone that is a major regulator of phosphate homeostasis. In this two-part review of disorders of phosphate homeostasis in children, part 1 covers the basics of mineral ion homeostasis and the roles of phosphate in skeletal biology. Part 1 includes phosphate-related disorders of mineralization for which overall circulating mineral ion homeostasis remains normal. Part 2 covers hypophosphatemic and hyperphosphatemic disorders, emphasizing, but not limited to, those related to increased and decreased FGF23 signaling, respectively.
Collapse
|
10
|
Couto AR, Parreira B, Power DM, Pinheiro L, Madruga Dias J, Novofastovski I, Eshed I, Sarzi-Puttini P, Pappone N, Atzeni F, Verlaan JJ, Kuperus J, Bieber A, Ambrosino P, Kiefer D, Khan MA, Mader R, Baraliakos X, Bruges-Armas J. Evidence for a genetic contribution to the ossification of spinal ligaments in Ossification of Posterior Longitudinal Ligament and Diffuse idiopathic skeletal hyperostosis: A narrative review. Front Genet 2022; 13:987867. [PMID: 36276944 PMCID: PMC9586552 DOI: 10.3389/fgene.2022.987867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/16/2022] [Indexed: 11/25/2022] Open
Abstract
Diffuse Idiopathic Skeletal Hyperostosis (DISH) and Ossification of the Posterior Longitudinal Ligament (OPLL) are common disorders characterized by the ossification of spinal ligaments. The cause for this ossification is currently unknown but a genetic contribution has been hypothesized. Over the last decade, many studies on the genetics of ectopic calcification disorders have been performed, mainly on OPLL. Most of these studies were based on linkage analysis and case control association studies. Animal models have provided some clues but so far, the involvement of the identified genes has not been confirmed in human cases. In the last few years, many common variants in several genes have been associated with OPLL. However, these associations have not been at definitive levels of significance and evidence of functional significance is generally modest. The current evidence suggests a multifactorial aetiopathogenesis for DISH and OPLL with a subset of cases showing a stronger genetic component.
Collapse
Affiliation(s)
- Ana Rita Couto
- Hospital de Santo Espirito da Ilha Terceira EPER, SEEBMO, Angra do Heroísmo, Portugal
- Comprehensive Health Research Centre, Hospital de Santo Espírito da Ilha Terceira, Lisbon, Portugal
| | - Bruna Parreira
- Hospital de Santo Espirito da Ilha Terceira EPER, SEEBMO, Angra do Heroísmo, Portugal
- Comprehensive Health Research Centre, Hospital de Santo Espírito da Ilha Terceira, Lisbon, Portugal
| | - Deborah M. Power
- University of Algarve, Center of Marine Science (CCMAR), Faro, Portugal
| | - Luís Pinheiro
- Hospital de Santo Espirito da Ilha Terceira EPER, Orthopedics Service, Angra do Heroísmo, Portugal
| | - João Madruga Dias
- Centro Hospitalar Do Medio Tejo EPE Unidade de Torres Novas, Rheumatology Department, Santarém, Portugal
- CHRC Campus Nova Medical School, EpiDoc Research Unit, CEDOC, Lisboa, Portugal
| | | | | | | | - Nicola Pappone
- Istituti Clinici Scientifici Maugeri IRCCS, Neuromotor Rehabilitation Unit of Telese Terme Institute, Pavia, Italy
| | - Fabiola Atzeni
- Universita Degli Studi di Messina, Rheumatology Unit, Clinical and Experimental Medicine, Messina, Italy
| | - Jorrit-Jan Verlaan
- University Medical Centre, Department of Orthopedics, Utrecht, Netherlands
| | | | - Amir Bieber
- Emek Medical Center, Rheumatology Unit, Afula, Israel
| | - Pasquale Ambrosino
- Istituti Clinici Scientifici Maugeri IRCCS, Cardiac Rehabilitation Unit of Telese Terme Institute, Pavia, Italy
| | - David Kiefer
- Ruhr-Universitat Bochum, Rheumazentrum Ruhrgebiet, Bochum, Germany
| | | | - Reuven Mader
- Emek Medical Center, Rheumatology Unit, Afula, Israel
- Rappaport Faculty of Medicine, Technion, Haifa, Israel
- Ruhr University Bochum, Rheumazentrum Ruhrgebiet, Herne, Germany
| | | | - Jácome Bruges-Armas
- Hospital de Santo Espirito da Ilha Terceira EPER, SEEBMO, Angra do Heroísmo, Portugal
- Comprehensive Health Research Centre, Hospital de Santo Espírito da Ilha Terceira, Lisbon, Portugal
| |
Collapse
|
11
|
Zimmerman K, Li X, von Kroge S, Stabach P, Lester ER, Chu EY, Srivastava S, Somerman MJ, Tommasini SM, Busse B, Schinke T, Carpenter TO, Oheim R, Braddock DT. Catalysis-Independent ENPP1 Protein Signaling Regulates Mammalian Bone Mass. J Bone Miner Res 2022; 37:1733-1749. [PMID: 35773783 PMCID: PMC9709593 DOI: 10.1002/jbmr.4640] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/09/2022] [Accepted: 06/17/2022] [Indexed: 11/06/2022]
Abstract
Biallelic ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) deficiency induces vascular/soft tissue calcifications in generalized arterial calcification of infancy (GACI), and low bone mass with phosphate-wasting rickets in GACI survivors (autosomal hypophosphatemic rickets type-2). ENPP1 haploinsufficiency induces early-onset osteoporosis and mild phosphate wasting in adults. Both conditions demonstrate the unusual combination of reduced accrual of skeletal mineral, yet excess and progressive heterotopic mineralization. ENPP1 is the only enzyme that generates extracellular pyrophosphate (PPi), a potent inhibitor of both bone and heterotopic mineralization. Life-threatening vascular calcification in ENPP1 deficiency is due to decreased plasma PPi; however, the mechanism by which osteopenia results is not apparent from an understanding of the enzyme's catalytic activity. To probe for catalysis-independent ENPP1 pathways regulating bone, we developed a murine model uncoupling ENPP1 protein signaling from ENPP1 catalysis, Enpp1T238A mice. In contrast to Enpp1asj mice, which lack ENPP1, Enpp1T238A mice have normal trabecular bone microarchitecture and favorable biomechanical properties. However, both models demonstrate low plasma Pi and PPi, increased fibroblast growth factor 23 (FGF23), and by 23 weeks, osteomalacia demonstrating equivalent phosphate wasting in both models. Reflecting findings in whole bone, calvarial cell cultures from Enpp1asj mice demonstrated markedly decreased calcification, elevated transcription of Sfrp1, and decreased nuclear β-catenin signaling compared to wild-type (WT) and Enpp1T238A cultures. Finally, the decreased calcification and nuclear β-catenin signaling observed in Enpp1asj cultures was restored to WT levels by knockout of Sfrp1. Collectively, our findings demonstrate that catalysis-independent ENPP1 signaling pathways regulate bone mass via the expression of soluble Wnt inhibitors such as secreted frizzled-related protein 1 (SFRP1), whereas catalysis dependent pathways regulate phosphate homeostasis through the regulation of plasma FGF23. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Kristin Zimmerman
- Department of Pathology, Yale University School of Medicine, New Haven Connecticut, 06510
| | - Xiaochen Li
- Department of Pathology, Yale University School of Medicine, New Haven Connecticut, 06510
| | - Simon von Kroge
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 22529 Hamburg, Germany
| | - Paul Stabach
- Department of Pathology, Yale University School of Medicine, New Haven Connecticut, 06510
| | - Ethan R. Lester
- Department of Pathology, Yale University School of Medicine, New Haven Connecticut, 06510
| | - Emily Y. Chu
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
- Department of General Dentistry, Operative Division, University of Maryland School of Dentistry, Baltimore, Maryland, 21202
| | - Shivani Srivastava
- Department of Pathology, Yale University School of Medicine, New Haven Connecticut, 06510
| | - Martha J. Somerman
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Steven M. Tommasini
- Department of Orthopædics and Rehabilitation, Yale University School of Medicine, New Haven Connecticut, 06510
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 22529 Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 22529 Hamburg, Germany
| | - Thomas O. Carpenter
- Department of Pediatrics at Yale University School of Medicine, New Haven Connecticut, 06510
| | - Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 22529 Hamburg, Germany
| | - Demetrios T. Braddock
- Department of Pathology, Yale University School of Medicine, New Haven Connecticut, 06510
| |
Collapse
|
12
|
Abstract
Inorganic phosphate (Pi) in the mammalian body is balanced by its influx and efflux through the intestines, kidneys, bones, and soft tissues, at which several sodium/Pi co-transporters mediate its active transport. Pi homeostasis is achieved through the complex counter-regulatory feedback balance between fibroblast growth factor 23 (FGF23), 1,25-dihydroxyvitamin D (1,25(OH)2D), and parathyroid hormone. FGF23, which is mainly produced by osteocytes in bone, plays a central role in Pi homeostasis and exerts its effects by binding to the FGF receptor (FGFR) and αKlotho in distant target organs. In the kidneys, the main target, FGF23 promotes the excretion of Pi and suppresses the production of 1,25(OH)2D. Deficient and excess FGF23 result in hyperphosphatemia and hypophosphatemia, respectively. FGF23-related hypophosphatemic rickets/osteomalacia include tumor-induced osteomalacia and various genetic diseases, such as X-linked hypophosphatemic rickets. Coverage by the national health insurance system in Japan for the measurement of FGF23 and the approval of burosumab, an FGF23-neutralizing antibody, have had a significant impact on the diagnosis and treatment of FGF23-related hypophosphatemic rickets/osteomalacia. Some of the molecules responsible for genetic hypophosphatemic rickets/osteomalacia are highly expressed in osteocytes and function as local regulators of FGF23 production. A number of systemic factors also regulate FGF23 levels. Although the mechanisms responsible for Pi sensing in mammals have not yet been elucidated in detail, recent studies have suggested the involvement of FGFR1. The further clarification of the mechanisms by which osteocytes detect Pi levels and regulate FGF23 production will lead to the development of better strategies to treat hyperphosphatemic and hypophosphatemic conditions.
Collapse
Affiliation(s)
- Toshimi Michigami
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Osaka 594-1101, Japan
| |
Collapse
|
13
|
Dsouza C, Moussa MS, Mikolajewicz N, Komarova SV. Extracellular ATP and its derivatives provide spatiotemporal guidance for bone adaptation to wide spectrum of physical forces. Bone Rep 2022; 17:101608. [PMID: 35992507 PMCID: PMC9385560 DOI: 10.1016/j.bonr.2022.101608] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/17/2022] Open
Abstract
ATP is a ubiquitous intracellular molecule critical for cellular bioenergetics. ATP is released in response to mechanical stimulation through vesicular release, small tears in cellular plasma membranes, or when cells are destroyed by traumatic forces. Extracellular ATP is degraded by ecto-ATPases to form ADP and eventually adenosine. ATP, ADP, and adenosine signal through purinergic receptors, including seven P2X ATP-gated cation channels, seven G-protein coupled P2Y receptors responsive to ATP and ADP, and four P1 receptors stimulated by adenosine. The goal of this review is to build a conceptual model of the role of different components of this complex system in coordinating cellular responses that are appropriate to the degree of mechanical stimulation, cell proximity to the location of mechanical injury, and time from the event. We propose that route and amount of ATP release depend on the scale of mechanical forces, ranging from vesicular release of small ATP boluses upon membrane deformation, to leakage of ATP through resealable plasma membrane tears, to spillage of cellular content due to destructive forces. Correspondingly, different P2 receptors responsive to ATP will be activated according to their affinity at the site of mechanical stimulation. ATP is a small molecule that readily diffuses through the environment, bringing the signal to the surrounding cells. ATP is also degraded to ADP which can stimulate a distinct set of P2 receptors. We propose that depending on the magnitude of mechanical forces and distance from the site of their application, ATP/ADP profiles will be different, allowing the relay of information about tissue level injury and proximity. Lastly, ADP is degraded to adenosine acting via its P1 receptors. The presence of large amounts of adenosine without ATP, indicates that an active source of ATP release is no longer present, initiating the transition to the recovery phase. This model consolidates the knowledge regarding the individual components of the purinergic system into a conceptual framework of choreographed responses to physical forces. Cellular bioenergetic molecule ATP is released when cell is mechanically stimulated. ATP release is proportional to the amount of cellular damage. ATP diffusion and transformation to ADP indicates the proximity to the damage. Purinergic receptors form a network choreographing cell response to physical forces. Complete transformation of ATP to adenosine initiates the recovery phase.
Collapse
Affiliation(s)
- Chrisanne Dsouza
- Department of Experimental Surgery, McGill University, Montreal, QC H3G 1A4, Canada
- Shriners Hospitals for Children- Canada, Montreal, QC H4A 0A9, Canada
| | - Mahmoud S. Moussa
- Shriners Hospitals for Children- Canada, Montreal, QC H4A 0A9, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| | - Nicholas Mikolajewicz
- Shriners Hospitals for Children- Canada, Montreal, QC H4A 0A9, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| | - Svetlana V. Komarova
- Department of Experimental Surgery, McGill University, Montreal, QC H3G 1A4, Canada
- Shriners Hospitals for Children- Canada, Montreal, QC H4A 0A9, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
- Corresponding author.
| |
Collapse
|
14
|
Theng EH, Brewer CC, Oheim R, Zalewski CK, King KA, Delsmann MM, Rolvien T, Gafni RI, Braddock DT, Jeffrey Kim H, Ferreira CR. Characterization of hearing-impairment in Generalized Arterial Calcification of Infancy (GACI). Orphanet J Rare Dis 2022; 17:273. [PMID: 35854274 PMCID: PMC9295326 DOI: 10.1186/s13023-022-02410-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/26/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND AND IMPORTANCE Hearing loss (HL) has been sporadically described, but not well characterized, in Generalized Arterial Calcification of Infancy (GACI), a rare disease in which pathological calcification typically presents in infancy. OBJECTIVES This study aims to describe the clinical audiologic and otologic features and potential etiology of hearing impairment in GACI and gain pathophysiological insight from a murine model of GACI. DESIGN Cross-sectional cohort study of individuals with GACI. Murine ossicle micromorphology of the ENPP1asj/asj mutant compared to wild-type. SETTING Clinical research hospital; basic science laboratory. PARTICIPANTS Nineteen individuals with GACI who met clinical, biochemical, and genetic criteria for diagnosis. MAIN OUTCOMES AND MEASURES Clinical, biochemical, and radiologic features associated with hearing status. RESULTS Pure-tone thresholds could be established in 15 (n = 30 ears) of the 19 patients who underwent audiological assessments. The prevalence of HL was 50% (15/30) of ears, with conductive HL in 80% and sensorineural HL in 20%. In terms of patients with HL (n = 8), seven patients had bilateral HL and one patient had unilateral HL. Degree of HL was mild to moderate for 87% of the 15 ears with hearing loss. Of those patients with sufficient pure-tone and middle ear function data, 80% (8/10) had audiometric configurations suggestive of ossicular chain dysfunction (OCD). Recurrent episodes of otitis media (ROM) requiring pressure-equalizing tube placement were common. In patients who underwent cranial CT, 54.5% (6/11) had auricular calcification. Quantitative backscattered electron imaging (qBEI) of murine ossicles supports an OCD component of auditory dysfunction in GACI, suggesting loss of ossicular osteocytes without initiation of bone remodeling. CONCLUSIONS AND RELEVANCE Hearing loss is common in GACI; it is most often conductive, and mild to moderate in severity. The etiology of HL is likely multifactorial, involving dysfunction of the ossicular chain and/or recurrent otitis media. Clinically, this study highlights the importance of early audiologic and otologic evaluation in persons with GACI. Novel findings of high rates of OCD and ROM may inform management, and in cases of unclear HL etiology, dedicated temporal bone imaging should be considered.
Collapse
Affiliation(s)
- Elizabeth H Theng
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Carmen C Brewer
- Audiology Unit, Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Ralf Oheim
- Martin Seitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christopher K Zalewski
- Audiology Unit, Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Kelly A King
- Audiology Unit, Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Maximillian M Delsmann
- Martin Seitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Trauma and Orthopaedic Surgery, Division of Orthopaedics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Rolvien
- Martin Seitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Trauma and Orthopaedic Surgery, Division of Orthopaedics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rachel I Gafni
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | | | - H Jeffrey Kim
- Office of Clinical Director, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA.
- Department of Otolaryngology-Head & Neck Surgery, District of Columbia, Georgetown University Hospital, Washington, USA.
| | - Carlos R Ferreira
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, 49 Convent Dr, Building 49, Room 4A38, Bethesda, MD, 20892, USA.
| |
Collapse
|
15
|
Kawai K, Sato Y, Kawakami R, Sakamoto A, Cornelissen A, Mori M, Ghosh S, Kutys R, Virmani R, Finn AV. Generalized Arterial Calcification of Infancy (GACI): Optimizing Care with a Multidisciplinary Approach. J Multidiscip Healthc 2022; 15:1261-1276. [PMID: 35677616 PMCID: PMC9167688 DOI: 10.2147/jmdh.s251861] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 04/22/2022] [Indexed: 11/23/2022] Open
Abstract
It is very unusual to see evidence of arterial calcification in infants and children, and when detected, genetic disorders of calcium metabolism should be suspected. Generalized arterial calcification of infancy (GACI) is a hereditary disease, which is characterized by severe arterial calcification of medium sized arteries, mostly involving the media with marked intimal proliferation and ectopic mineralization of the extravascular tissues. It is caused by inactivating variants in genes encoding either ENPP1, in a majority of cases (70–75%), or ABCC6, in a minority (9–10%). Despite similar histologic appearances between ENPP1 and ABCC6 deficiencies, including arterial calcification, organ calcification, and cardiovascular calcification, mortality is higher in subjects carrying the ENPP1 versus ABCC6 variants (40% vs 10%, respectively). Overall mortality in individuals with GACI is high (55%) before the age of 6 months, with 24.4% dying in utero or being stillborn. Rare cases show spontaneous regression with age, while others who survive into adulthood often manifest musculoskeletal complications (osteoarthritis and interosseous membrane ossification), enthesis mineralization, and cervical spine fusion. Despite recent advances in the understanding of the genetic mechanisms underlying this disease, there is still no ideal therapy for the resolution of vascular calcification in GACI. Although bisphosphonates with anti-calcification properties have been commonly used for the treatment of CAGI, their benefit is controversial, with favorable results reported at one year and questionable benefit with delayed initiation of treatment. Enzyme replacement therapy with administration of recombinant form of ENPP1 prevents calcification and mortality, improves hypertension and cardiac function, and prevents intimal proliferation and osteomalacia in mouse models of ENPP1 deficiency. Therefore, newer treatments targeting genes are on the horizon. In this article, we review up to date knowledge of the understanding of GACI, its clinical, pathologic, and etiologic understanding and treatment in support of more comprehensive care of GACI patients.
Collapse
Affiliation(s)
| | - Yu Sato
- CVPath Institute, Gaithersburg, MD, USA
| | | | | | | | | | | | | | | | - Aloke V Finn
- CVPath Institute, Gaithersburg, MD, USA
- University of Maryland, School of Medicine, Baltimore, MD, USA
- Correspondence: Aloke V Finn, 19 Firstfield Road, Gaithersburg, MD, 20878, USA, Tel +301.208.3570, Fax +301.208.3745, Email
| |
Collapse
|
16
|
Ye J, Calvo IA, Cenzano I, Vilas A, Martinez-de-Morentin X, Lasaga M, Alignani D, Paiva B, Viñado AC, San Martin-Uriz P, Romero JP, Quilez Agreda D, Miñana Barrios M, Sancho-González I, Todisco G, Malcovati L, Planell N, Saez B, Tegner JN, Prosper F, Gomez-Cabrero D. Deconvolution of the hematopoietic stem cell microenvironment reveals a high degree of specialization and conservation. iScience 2022; 25:104225. [PMID: 35494238 PMCID: PMC9046238 DOI: 10.1016/j.isci.2022.104225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/14/2022] [Accepted: 04/05/2022] [Indexed: 11/28/2022] Open
Abstract
Understanding the regulation of normal and malignant human hematopoiesis requires comprehensive cell atlas of the hematopoietic stem cell (HSC) regulatory microenvironment. Here, we develop a tailored bioinformatic pipeline to integrate public and proprietary single-cell RNA sequencing (scRNA-seq) datasets. As a result, we robustly identify for the first time 14 intermediate cell states and 11 stages of differentiation in the endothelial and mesenchymal BM compartments, respectively. Our data provide the most comprehensive description to date of the murine HSC-regulatory microenvironment and suggest a higher level of specialization of the cellular circuits than previously anticipated. Furthermore, this deep characterization allows inferring conserved features in human, suggesting that the layers of microenvironmental regulation of hematopoiesis may also be shared between species. Our resource and methodology is a stepping-stone toward a comprehensive cell atlas of the BM microenvironment.
Collapse
Affiliation(s)
- Jin Ye
- Bioscience Program, Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology KAUST, Thuwal 23955, Saudi Arabia
| | - Isabel A. Calvo
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Itziar Cenzano
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
| | - Amaia Vilas
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Xabier Martinez-de-Morentin
- Navarrabiomed, ComplejoHospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, 31008 Navarra, Spain
| | - Miren Lasaga
- Navarrabiomed, ComplejoHospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, 31008 Navarra, Spain
| | - Diego Alignani
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Bruno Paiva
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Ana C. Viñado
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Patxi San Martin-Uriz
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
| | - Juan P. Romero
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
| | | | | | | | - Gabriele Todisco
- Department of Molecular Medicine, University of Pavia & Unit of Precision Hematology Oncology, IRCCS S. Matteo Hospital Foundation, 27100 Pavia, Italy
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Luca Malcovati
- Department of Molecular Medicine, University of Pavia & Unit of Precision Hematology Oncology, IRCCS S. Matteo Hospital Foundation, 27100 Pavia, Italy
| | - Nuria Planell
- Navarrabiomed, ComplejoHospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, 31008 Navarra, Spain
| | - Borja Saez
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Jesper N. Tegner
- Bioscience Program, Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology KAUST, Thuwal 23955, Saudi Arabia
- Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Stockholm, Sweden
- Computer, Electrical, and Mathematical Sciences and Engineering Division (CEMSE), King Abdullah University of Science and Technology KAUST, Thuwal 23955, Saudi Arabia
- Bioengineering Program, Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology KAUST, Thuwal 23955, Saudi Arabia
| | - Felipe Prosper
- Universidad de Navarra, CIMA, Hematology-Oncology Program, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Navarra, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
- Service of Hematology and Cell Therapy, Clínica Universidad de Navarra; CCUN, Pamplona, Navarra, 31008; Spain
| | - David Gomez-Cabrero
- Bioscience Program, Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology KAUST, Thuwal 23955, Saudi Arabia
- Navarrabiomed, ComplejoHospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, 31008 Navarra, Spain
- Department of Medicine, Centre for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Stockholm, Sweden
- Centre for Host Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College, London WC2R 2LS, UK
- Bioengineering Program, Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology KAUST, Thuwal 23955, Saudi Arabia
| |
Collapse
|
17
|
Höppner J, Kornak U, Sinningen K, Rutsch F, Oheim R, Grasemann C. Autosomal recessive hypophosphatemic rickets type 2 (ARHR2) due to ENPP1-deficiency. Bone 2021; 153:116111. [PMID: 34252603 DOI: 10.1016/j.bone.2021.116111] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 12/25/2022]
Abstract
Awareness for hypophosphatemic rickets has increased in the last years, based on the availability of specific medical treatments. Autosomal recessive hypophosphatemic rickets type 2 (ARHR2) is a rare form of hypophosphatemic rickets, which is known to develop in survivors of generalized arterial calcification of infancy (GACI). Both disorders are based on a deficiency of ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) and present with a high clinical variability and a lack of a phenotype-genotype association. ARHR2 is characterized by phosphate wasting due to elevated fibroblast growth factor 23 (FGF23) levels and might represent a response of the organism to minimize ectopic calcification in individuals with ENPP1-deficiency. This report reviews the recent clinical and preclinical data on this ultra-rare disease in childhood.
Collapse
Affiliation(s)
- Jakob Höppner
- Center for Rare Diseases Ruhr CeSER, Ruhr-University Bochum and Witten/Herdecke University, Germany; Department of Pediatrics, St.-Josef Hospital Bochum, Ruhr-University Bochum, Bochum, Germany
| | - Uwe Kornak
- Institute for Human Genetics, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Kathrin Sinningen
- Department of Pediatrics, St.-Josef Hospital Bochum, Ruhr-University Bochum, Bochum, Germany
| | - Frank Rutsch
- Department of General Pediatrics, Münster University Children's Hospital, Münster, Germany
| | - Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Corinna Grasemann
- Center for Rare Diseases Ruhr CeSER, Ruhr-University Bochum and Witten/Herdecke University, Germany; Department of Pediatrics, St.-Josef Hospital Bochum, Ruhr-University Bochum, Bochum, Germany.
| |
Collapse
|
18
|
Enzymatic Approach in Calcium Phosphate Biomineralization: A Contribution to Reconcile the Physicochemical with the Physiological View. Int J Mol Sci 2021; 22:ijms222312957. [PMID: 34884758 PMCID: PMC8657759 DOI: 10.3390/ijms222312957] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 01/22/2023] Open
Abstract
Biomineralization is the process by which organisms produce hard inorganic matter from soft tissues with outstanding control of mineral deposition in time and space. For this purpose, organisms deploy a sophisticated "toolkit" that has resulted in significant evolutionary innovations, for which calcium phosphate (CaP) is the biomineral selected for the skeleton of vertebrates. While CaP mineral formation in aqueous media can be investigated by studying thermodynamics and kinetics of phase transitions in supersaturated solutions, biogenic mineralization requires coping with the inherent complexity of biological systems. This mainly includes compartmentalization and homeostatic processes used by organisms to regulate key physiological factors, including temperature, pH and ion concentration. A detailed analysis of the literature shows the emergence of two main views describing the mechanism of CaP biomineralization. The first one, more dedicated to the study of in vivo systems and supported by researchers in physiology, often involves matrix vesicles (MVs). The second one, more investigated by the physicochemistry community, involves collagen intrafibrillar mineralization particularly through in vitro acellular models. Herein, we show that there is an obvious need in the biological systems to control both where and when the mineral forms through an in-depth survey of the mechanism of CaP mineralization. This necessity could gather both communities of physiologists and physicochemists under a common interest for an enzymatic approach to better describe CaP biomineralization. Both homogeneous and heterogeneous enzymatic catalyses are conceivable for these systems, and a few preliminary promising results on CaP mineralization for both types of enzymatic catalysis are reported in this work. Through them, we aim to describe the relevance of our point of view and the likely findings that could be obtained when adding an enzymatic approach to the already rich and creative research field dealing with CaP mineralization. This complementary approach could lead to a better understanding of the biomineralization mechanism and inspire the biomimetic design of new materials.
Collapse
|
19
|
Bourne LE, Patel JJ, Davies BK, Neven E, Verhulst A, D'Haese PC, Wheeler-Jones CPD, Orriss IR. N-acetylcysteine (NAC) differentially affects arterial medial calcification and bone formation: The role of l-cysteine and hydrogen sulphide. J Cell Physiol 2021; 237:1070-1086. [PMID: 34658034 DOI: 10.1002/jcp.30605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 12/29/2022]
Abstract
Arterial medial calcification (AMC) is the deposition of calcium phosphate in the arteries. AMC is widely thought to share similarities with physiological bone formation; however, emerging evidence suggests several key differences between these processes. N-acetylcysteine (NAC) displays antioxidant properties and can generate hydrogen sulphide (H2 S) and glutathione (GSH) from its deacetylation to l-cysteine. This study found that NAC exerts divergent effects in vitro, increasing osteoblast differentiation and bone formation by up to 5.5-fold but reducing vascular smooth muscle cell (VSMC) calcification and cell death by up to 80%. In vivo, NAC reduced AMC in a site-specific manner by 25% but had no effect on the bone. The actions of l-cysteine and H2 S mimicked those of NAC; however, the effects of H2 S were much less efficacious than NAC and l-cysteine. Pharmacological inhibition of H2 S-generating enzymes did not alter the actions of NAC or l-cysteine; endogenous production of H2 S was also unaffected. In contrast, NAC and l-cysteine increased GSH levels in calcifying VSMCs and osteoblasts by up to 3-fold. This suggests that the beneficial actions of NAC are likely to be mediated via the breakdown of l-cysteine and the subsequent GSH generation. Together, these data show that while the molecular mechanisms driving the actions of NAC appear similar, the downstream effects on cell function differ significantly between osteoblasts and calcifying VSMCs. The ability of NAC to exert these differential actions further supports the notion that there are differences between the development of pathological AMC and physiological bone formation. NAC could represent a therapeutic option for treating AMC without exerting negative effects on bone.
Collapse
Affiliation(s)
- Lucie E Bourne
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Jessal J Patel
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Bethan K Davies
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Ellen Neven
- Department of Biomedical Sciences, Laboratory of Pathophysiology, University of Antwerp, Antwerp, Belgium
| | - Anja Verhulst
- Department of Biomedical Sciences, Laboratory of Pathophysiology, University of Antwerp, Antwerp, Belgium
| | - Patrick C D'Haese
- Department of Biomedical Sciences, Laboratory of Pathophysiology, University of Antwerp, Antwerp, Belgium
| | | | - Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| |
Collapse
|
20
|
Cheng Z, O'Brien K, Howe J, Sullivan C, Schrier D, Lynch A, Jungles S, Sabbagh Y, Thompson D. INZ-701 Prevents Ectopic Tissue Calcification and Restores Bone Architecture and Growth in ENPP1-Deficient Mice. J Bone Miner Res 2021; 36:1594-1604. [PMID: 33900645 DOI: 10.1002/jbmr.4315] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 04/08/2021] [Accepted: 04/17/2021] [Indexed: 12/20/2022]
Abstract
Ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) is the major enzyme that cleaves extracellular adenosine triphosphate (ATP) to generate pyrophosphate (PPi), an inorganic metabolite with potent anticalcification activity. Loss-of-function mutations cause hypopyrophosphatemia and lead to a state of ENPP1 deficiency, which has an acute infantile phase known as generalized arterial calcification of infancy (GACI) and a pediatric to adult phase known as autosomal-recessive hypophosphatemic rickets type 2 (ARHR2). ENPP1 deficiency manifests as ectopic calcification of multiple tissues, neointimal proliferation, premature mortality, impaired growth, and bone deformities. INZ-701, a human ENPP1-Fc protein, is in clinical development as an enzyme replacement therapy for the treatment of ENPP1 deficiency. The pharmacokinetic and pharmacodynamic profile and therapeutic effect of INZ-701 were investigated in Enpp1asj/asj mice, a murine model of ENPP1 deficiency. Enpp1asj/asj mice have undetectable plasma PPi, lower plasma phosphate, and higher FGF23 levels compared with wild-type (WT) mice. Enpp1asj/asj mice on the acceleration diet, containing high phosphate and low magnesium, quickly develop clinical signs, including dehydration, rough hair coat, pinned ears, stiffed legs, and hunched back. Enpp1asj/asj mice treated with vehicle had aforementioned clinical signs plus severe ectopic calcification in multiple tissues and bone defects, characteristics of the clinical phenotype observed in GACI and ARHR2 patients. Our results showed a durable PPi response for more than 3 days after a single dose of INZ-701. Treatment of ENPP1-deficient mice every other day with INZ-701 for 8 weeks restored circulating levels of PPi, prevented pathological calcification in all the tested organs, restored growth parameters, corrected bone defects, improved clinical signs, and decreased mortality in Enpp1asj/asj mice, demonstrating the potential of INZ-701 to treat ENPP1 deficiency. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
|
21
|
Roberts FL, Rashdan NA, Phadwal K, Markby GR, Dillon S, Zoll J, Berger J, Milne E, Orriss IR, Karsenty G, Le Saux O, Morton NM, Farquharson C, MacRae VE. Osteoblast-specific deficiency of ectonucleotide pyrophosphatase or phosphodiesterase-1 engenders insulin resistance in high-fat diet fed mice. J Cell Physiol 2021; 236:4614-4624. [PMID: 33305372 PMCID: PMC9665351 DOI: 10.1002/jcp.30194] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 11/14/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022]
Abstract
Supraphysiological levels of the osteoblast-enriched mineralization regulator ectonucleotide pyrophosphatase or phosphodiesterase-1 (NPP1) is associated with type 2 diabetes mellitus. We determined the impact of osteoblast-specific Enpp1 ablation on skeletal structure and metabolic phenotype in mice. Female, but not male, 6-week-old mice lacking osteoblast NPP1 expression (osteoblast-specific knockout [KO]) exhibited increased femoral bone volume or total volume (17.50% vs. 11.67%; p < .01), and reduced trabecular spacing (0.187 vs. 0.157 mm; p < .01) compared with floxed (control) mice. Furthermore, an enhanced ability of isolated osteoblasts from the osteoblast-specific KO to calcify their matrix in vitro compared to fl/fl osteoblasts was observed (p < .05). Male osteoblast-specific KO and fl/fl mice showed comparable glucose and insulin tolerance despite increased levels of insulin-sensitizing under-carboxylated osteocalcin (195% increase; p < .05). However, following high-fat-diet challenge, osteoblast-specific KO mice showed impaired glucose and insulin tolerance compared with fl/fl mice. These data highlight a crucial local role for osteoblast NPP1 in skeletal development and a secondary metabolic impact that predominantly maintains insulin sensitivity.
Collapse
Affiliation(s)
- Fiona L. Roberts
- Functional Genetics and Development, The Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Nabil A. Rashdan
- Functional Genetics and Development, The Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Kanchan Phadwal
- Functional Genetics and Development, The Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Greg R. Markby
- Functional Genetics and Development, The Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Scott Dillon
- Functional Genetics and Development, The Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Janna Zoll
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Julian Berger
- Department of Genetics and Development, Columbia University Medical Center, New York, New York, USA
| | - Elspeth Milne
- Functional Genetics and Development, The Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Isabel R. Orriss
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| | - Gerard Karsenty
- Department of Genetics and Development, Columbia University Medical Center, New York, New York, USA
| | - Olivier Le Saux
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Nicholas M. Morton
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, The College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Colin Farquharson
- Functional Genetics and Development, The Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Vicky E. MacRae
- Functional Genetics and Development, The Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Midlothian, UK
| |
Collapse
|
22
|
Ferreira CR, Kavanagh D, Oheim R, Zimmerman K, Stürznickel J, Li X, Stabach P, Rettig RL, Calderone L, MacKichan C, Wang A, Hutchinson HA, Nelson T, Tommasini SM, von Kroge S, Fiedler IA, Lester ER, Moeckel GW, Busse B, Schinke T, Carpenter TO, Levine MA, Horowitz MC, Braddock DT. Response of the ENPP1-Deficient Skeletal Phenotype to Oral Phosphate Supplementation and/or Enzyme Replacement Therapy: Comparative Studies in Humans and Mice. J Bone Miner Res 2021; 36:942-955. [PMID: 33465815 PMCID: PMC8739051 DOI: 10.1002/jbmr.4254] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 01/11/2021] [Accepted: 01/15/2021] [Indexed: 12/14/2022]
Abstract
Inactivating mutations in human ecto-nucleotide pyrophosphatase/phosphodiesterase-1 (ENPP1) may result in early-onset osteoporosis (EOOP) in haploinsufficiency and autosomal recessive hypophosphatemic rickets (ARHR2) in homozygous deficiency. ARHR2 patients are frequently treated with phosphate supplementation to ameliorate the rachitic phenotype, but elevating plasma phosphorus concentrations in ARHR2 patients may increase the risk of ectopic calcification without increasing bone mass. To assess the risks and efficacy of conventional ARHR2 therapy, we performed comprehensive evaluations of ARHR2 patients at two academic medical centers and compared their skeletal and renal phenotypes with ENPP1-deficient Enpp1asj/asj mice on an acceleration diet containing high phosphate treated with recombinant murine Enpp1-Fc. ARHR2 patients treated with conventional therapy demonstrated improvements in rickets, but all adults and one adolescent analyzed continued to exhibit low bone mineral density (BMD). In addition, conventional therapy was associated with the development of medullary nephrocalcinosis in half of the treated patients. Similar to Enpp1asj/asj mice on normal chow and to patients with mono- and biallelic ENPP1 mutations, 5-week-old Enpp1asj/asj mice on the high-phosphate diet exhibited lower trabecular bone mass, reduced cortical bone mass, and greater bone fragility. Treating the Enpp1asj/asj mice with recombinant Enpp1-Fc protein between weeks 2 and 5 normalized trabecular bone mass, normalized or improved bone biomechanical properties, and prevented the development of nephrocalcinosis and renal failure. The data suggest that conventional ARHR2 therapy does not address low BMD inherent in ENPP1 deficiency, and that ENPP1 enzyme replacement may be effective for correcting low bone mass in ARHR2 patients without increasing the risk of nephrocalcinosis. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Carlos R Ferreira
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dillon Kavanagh
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristin Zimmerman
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Julian Stürznickel
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Xiaofeng Li
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Paul Stabach
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - R Luke Rettig
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Logan Calderone
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Colin MacKichan
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Aaron Wang
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Hunter A Hutchinson
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Tracy Nelson
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Steven M Tommasini
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Simon von Kroge
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Imke Ak Fiedler
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ethan R Lester
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Gilbert W Moeckel
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas O Carpenter
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Michael A Levine
- Department of Pediatrics, University of Pennsylvania Perlman School of Medicine, Philadelphia, PA, USA.,Division of Endocrinology and Diabetes and Center for Bone Health, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mark C Horowitz
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
23
|
Magnesium whitlockite - omnipresent in pathological mineralisation of soft tissues but not a significant inorganic constituent of bone. Acta Biomater 2021; 125:72-82. [PMID: 33610767 DOI: 10.1016/j.actbio.2021.02.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/26/2021] [Accepted: 02/12/2021] [Indexed: 01/03/2023]
Abstract
Whitlockite is a calcium phosphate that was first identified in minerals collected from the Palermo Quarry, New Hampshire. The terms magnesium whitlockite [Mg-whitlockite; Ca18Mg2(HPO4)2(PO4)12] and beta-tricalcium phosphate [β-TCP; β-Ca3(PO4)2] are often used interchangeably since Mg-whitlockite is not easily distinguished from β-Ca3(PO4)2 by powder X-ray diffraction although their crystalline structures differ significantly. Being both osteoconductive and bioresorbable, Mg-whitlockite is pursued as a synthetic bone graft substitute. In recent years, advances in development of synthetic Mg-whitlockite have been accompanied by claims that Mg-whitlockite is the second most abundant inorganic constituent of bone, occupying as much as 20-35 wt% of the inorganic fraction. To find evidence in support of this notion, this review presents an exhaustive summary of Mg-whitlockite identification in biological tissues. Mg-whitlockite is mainly found in association with pathological mineralisation of various soft tissues and dental calculus, and occasionally with enamel and dentine. With the exception of high-temperature treated tumoural calcified deposits around interphalangeal and metacarpal joints and rhomboidal Mg-whitlockite crystals in post-apoptotic osteocyte lacunae in human alveolar bone, this unusual mineral has never been detected in the extracellular matrix of mammalian bone. Characterisation techniques capable of unequivocally distinguishing between different calcium phosphate phases, such as high-resolution imaging, crystallography, and/or spectroscopy have exclusively identified bone mineral as poorly crystalline, ion-substituted, carbonated apatite. The idea that Mg-whitlockite is a significant constituent of bone mineral remains unsubstantiated. Contrary to claims that such biomaterials represent a bioinspired/biomimetic approach to bone repair, Mg-whitlockite remains, exclusively, a pathological biomineral. STATEMENT OF SIGNIFICANCE: Magnesium whitlockite (Mg-whitlockite) is a unique calcium phosphate that typically features in pathological calcification of soft tissues; however, an alarming trend emerging in the synthetic bioceramics community claims that Mg-whitlockite occupies 20-35 wt% of bone mineral and therefore synthetic Mg-whitlockite represents a biomimetic approach towards bone regeneration. By providing an overview of Mg-whitlockite detection in biological tissues and scrutinising a diverse cross-section of literature relevant to bone composition analysis, this review concludes that Mg-whitlockite is exclusively a pathological biomineral, and having never been reported in bone extracellular matrix, Mg-whitlockite does not constitute a biomimetic strategy for bone repair.
Collapse
|
24
|
Extracellular purines and bone homeostasis. Biochem Pharmacol 2021; 187:114425. [PMID: 33482152 DOI: 10.1016/j.bcp.2021.114425] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 12/12/2022]
Abstract
Maintenance of a healthy skeleton is highly dependent on an intricate regulation of bone metabolism, as changes in the balance between bone formation and bone resorption leads to bone loss, bone fragility and ultimately bone fractures. During the last three decades it has become increasingly evident that physiological release of purines in the extracellular space is imperative for bone homeostasis and is orchestrated via the network of purinoceptors. Adenosine derivatives are released locally in the skeleton either by the bone forming osteoblasts or the bone degrading osteoclasts actioned directly by processes like mechanical loading and indirectly by systemic hormones. Adenosine derivatives directly affect the bone cells by their action on the membranal receptors or have co-stimulatory actions with bone active hormones such as parathyroid hormone or the gut hormones. Any deviations leading to increased levels of extracellular adenosine derivatives in the bone tissue such as in pathologic situations, trigger complex pathways with opposing effects on tissue health as presented by studies involving a range of model organisms. Pathological conditions where skeletal purinergic signaling is affected are following tissue injury like microdamage and macroscopic fractures; and during inflammatory processes where nucleotides and nucleosides play an important part in the pathophysiological skeletal response. Moreover, adenosine derivatives also play an important role in the interaction between malignant cells and bone cells in several types of cancers involving the skeleton, such as but not limited to multiple myeloma and bone osteolysis. Much knowledge has been gained over the last decades. The net- resulting phenotype of adenosine derivatives in bone (including the ratio of ATP to Adenosine) is highly dependent on CD39 and CD73 enzymes together with the expression and activity of the specific receptors. Thus, each component is important in the physiological and pathophysiological processes in bone. Promising perspectives await in the future in treating skeletal disorders with medications targeting the individual components of the purinergic signaling pathway.
Collapse
|
25
|
Niada S, Giannasi C, Magagnotti C, Andolfo A, Brini AT. Proteomic analysis of extracellular vesicles and conditioned medium from human adipose-derived stem/stromal cells and dermal fibroblasts. J Proteomics 2020; 232:104069. [PMID: 33309826 DOI: 10.1016/j.jprot.2020.104069] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/23/2020] [Accepted: 11/29/2020] [Indexed: 12/19/2022]
Abstract
Conditioned medium (CM) and extracellular vesicles (EV) from Adipose-derived Stem/stromal cells (ASC) and Dermal fibroblasts (DF) represent promising tools for therapeutic applications. Which one should be preferred is still under debate and no direct comparison of their proteome has been reported yet. Here, we apply quantitative proteomics to explore the protein composition of CM and EV from the two cell types. Data are available via ProteomeXchange (identifier PXD020219). We identified 1977 proteins by LC-MS/MS proteomic analysis. Unsupervised clustering analysis and PCA recognized CM and EV as separate groups. We identified 68 and 201 CM and EV specific factors. CM were enriched in proteins of endoplasmic reticulum, Golgi apparatus and lysosomes, whereas EV contained a large amount of GTPases, ribosome and translation factors. The analysis of ASC and DF secretomes revealed the presence of cell type-specific proteins. ASC-CM and -EV carried factors involved in ECM organization and immunological regulation, respectively. Conversely, DF-CM and -EV were enriched in epithelium development associated factors and -EV in Wnt signaling factors. In conclusion, this analysis provides evidence of a different protein composition between CM and EV and of the presence of cell type-specific bioactive mediators suggesting their specific future use as advanced therapy medicinal products. SIGNIFICANCE: The use of cell secretome presents several advantages over cell therapy such as the lower risks associated to the administration step and the avoidance of any potential risk of malignant transformation. The main secretome preparations consist in concentrated conditioned medium (CM) and extracellular vesicles (EV). Both of them showed well-documented therapeutic potentials. However, it is still not clear in which case it should be better to use one preparation over the other and an exhaustive comparison between their proteome has not been performed yet. The choice of the cell source is another relevant aspect that still needs to be addressed. In order to shed light on these questions we explored the protein composition of CM and EV obtained from Adipose-derived Stem/stromal Cells (ASC) and Dermal Fibroblasts (DF), by a comprehensive quantitative proteomics approach. The analysis showed a clear distinction between CM and EV proteome. CM were enriched in proteins of endoplasmic reticulum, Golgi apparatus and lysosomes, whereas EV contained a large amount of GTPases, ribosome and translation-related factors. Furthermore, the analysis of ASC and DF secretomes revealed specific biological processes for the different cell products. ASC secretome presented factors involved in ECM organization (hyaluronan and glycosaminoglycan metabolism) and immunological regulation (e.g. macrophage and IkB/NFkB signaling regulation), respectively. On the other hand, DF-CM and -EV were both enriched in epithelium development associated factors, whilst DF-CM in proteins involved in cellular processes regulation and -EV in Wnt signaling factors. In conclusion, our study shed a light on the different protein composition of CM and EV of two promising cell types, spanning from basic processes involved in secretion to specific pathways supporting their therapeutic potential and their possible future use as advanced therapy medicinal products.
Collapse
Affiliation(s)
| | | | - Cinzia Magagnotti
- Proteomics and Metabolomics Facility (ProMeFa), IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Annapaola Andolfo
- Proteomics and Metabolomics Facility (ProMeFa), IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Anna Teresa Brini
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy; Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
26
|
Gabaton N, Kannu P, Pope E, Shugar A, Lara-Corrales I. A novel ENPP1 mutation identified in a multigenerational family affected by Cole disease. Pediatr Dermatol 2020; 37:868-871. [PMID: 32598042 DOI: 10.1111/pde.14222] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/23/2020] [Accepted: 05/10/2020] [Indexed: 12/12/2022]
Abstract
Cole disease is a rare autosomal dominant genodermatosis with only five cases published in literature since its first description in 1976. We report a case of a 3-year-old boy of Italian ancestry who presented with hypopigmented skin patches on the upper extremities and multiple yellowish, firm papules and small plaques on his palms and soles. There were similar findings in the family, extending back at least four generations. Whole exome sequence analysis revealed a novel variant of the ENPP1 gene mutation, which has not been previously reported to be associated with Cole disease. Although there is no extracutaneous involvement associated with this condition, accurate diagnosis and variant identification is nevertheless important so that appropriate medical and genetic counseling can be offered to affected individuals and their at-risk relatives.
Collapse
Affiliation(s)
- Niña Gabaton
- Section of Dermatology, Division of Paediatric Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Peter Kannu
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Elena Pope
- Section of Dermatology, Division of Paediatric Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Andrea Shugar
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Irene Lara-Corrales
- Section of Dermatology, Division of Paediatric Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
27
|
Calcinosis in Systemic Sclerosis: Updates in Pathophysiology, Evaluation, and Treatment. Curr Rheumatol Rep 2020; 22:73. [DOI: 10.1007/s11926-020-00951-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
28
|
Chu EY, Vo TD, Chavez MB, Nagasaki A, Mertz EL, Nociti FH, Aitken SF, Kavanagh D, Zimmerman K, Li X, Stabach PR, Braddock DT, Millán JL, Foster BL, Somerman MJ. Genetic and pharmacologic modulation of cementogenesis via pyrophosphate regulators. Bone 2020; 136:115329. [PMID: 32224162 PMCID: PMC7482720 DOI: 10.1016/j.bone.2020.115329] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 11/27/2022]
Abstract
Pyrophosphate (PPi) serves as a potent and physiologically important regulator of mineralization, with systemic and local concentrations determined by several key regulators, including: tissue-nonspecific alkaline phosphatase (ALPL gene; TNAP protein), the progressive ankylosis protein (ANKH; ANK), and ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1; ENPP1). Results to date have indicated important roles for PPi in cementum formation, and we addressed several gaps in knowledge by employing genetically edited mouse models where PPi metabolism was disrupted and pharmacologically modulating PPi in a PPi-deficient mouse model. We demonstrate that acellular cementum growth is inversely proportional to PPi levels, with reduced cementum in Alpl KO (increased PPi levels) mice and excess cementum in Ank KO mice (decreased PPi levels). Moreover, simultaneous ablation of Alpl and Ank results in reestablishment of functional cementum in dKO mice. Additional reduction of PPi by dual deletion of Ank and Enpp1 does not further increase cementogenesis, and PDL space is maintained in part through bone modeling/remodeling by osteoclasts. Our results provide insights into cementum formation and expand our knowledge of how PPi regulates cementum. We also demonstrate for the first time that pharmacologic manipulation of PPi through an ENPP1-Fc fusion protein can regulate cementum growth, supporting therapeutic interventions targeting PPi metabolism.
Collapse
Affiliation(s)
- E Y Chu
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA.
| | - T D Vo
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - M B Chavez
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - A Nagasaki
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - E L Mertz
- National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - F H Nociti
- Department of Prosthodontics & Periodontics, State University of Campinas, Piracicaba Dental School, Piracicaba, São Paulo, Brazil
| | - S F Aitken
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - D Kavanagh
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - K Zimmerman
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - X Li
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - P R Stabach
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - D T Braddock
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - J L Millán
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - B L Foster
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - M J Somerman
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
29
|
Boyce AM, Gafni RI, Ferreira CR. Generalized Arterial Calcification of Infancy: New Insights, Controversies, and Approach to Management. Curr Osteoporos Rep 2020; 18:232-241. [PMID: 32172442 PMCID: PMC9506683 DOI: 10.1007/s11914-020-00577-4] [Citation(s) in RCA: 20] [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] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW This review summarizes current understanding of generalized arterial calcification of infancy (GACI), emphasizing pathophysiology, clinical presentation, and approaches and controversies in management. RECENT FINDINGS Identification of causative ENPP1 mutations revealed that GACI arises from deficiencies in inorganic pyrophosphate (leading to calcifications) and adenosine monophosphate (leading to intimal proliferation). Identification of genotypic and phenotypic overlap with pseudoxanthoma elasticum and autosomal recessive hypophosphatemic rickets further advanced understanding of GACI as a complex, multisystemic disease. Clinical data is limited to small, retrospective samples; it is therefore unknown whether commonly used medications, such as bisphosphonates and hypophosphatemia treatment, are therapeutic or potentially harmful. ENPP1-Fc replacement represents a promising approach warranting further study. Knowledge gaps in natural history place clinicians at high risk of assigning causality to interventions that are correlated with changes in clinical status. There is thus a critical need for improved natural history studies to develop and test targeted therapies.
Collapse
Affiliation(s)
- Alison M Boyce
- Skeletal Diseases and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive Room 218 MSC 4320, Bethesda, MD, 20892, USA.
| | - Rachel I Gafni
- Skeletal Diseases and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive Room 218 MSC 4320, Bethesda, MD, 20892, USA
| | - Carlos R Ferreira
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
30
|
Orriss IR. Extracellular pyrophosphate: The body's "water softener". Bone 2020; 134:115243. [PMID: 31954851 DOI: 10.1016/j.bone.2020.115243] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/08/2020] [Accepted: 01/16/2020] [Indexed: 12/18/2022]
Abstract
Extracellular pyrophosphate (ePPi) was first identified as a key endogenous inhibitor of mineralisation in the 1960's by Fleisch and colleagues. The main source of ePPi seems to be extracellular ATP which is continually released from cells in a controlled way. ATP is rapidly broken down by enzymes including ecto-nucleotide pyrophosphatase/phosphodiesterases to produce ePPi. The major function of ePPi is to directly inhibit hydroxyapatite formation and growth meaning that this simple molecule acts as the body's own "water softener". However, studies have also shown that ePPi can influence gene expression and regulate its own production and breakdown. This review will summarise our current knowledge of ePPi metabolism and how it acts to prevent pathological soft tissue calcification and regulate physiological bone mineralisation.
Collapse
Affiliation(s)
- Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK.
| |
Collapse
|
31
|
Oheim R, Zimmerman K, Maulding ND, Stürznickel J, von Kroge S, Kavanagh D, Stabach PR, Kornak U, Tommasini SM, Horowitz MC, Amling M, Thompson D, Schinke T, Busse B, Carpenter TO, Braddock DT. Human Heterozygous ENPP1 Deficiency Is Associated With Early Onset Osteoporosis, a Phenotype Recapitulated in a Mouse Model of Enpp1 Deficiency. J Bone Miner Res 2020; 35:528-539. [PMID: 31805212 PMCID: PMC7184798 DOI: 10.1002/jbmr.3911] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/27/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022]
Abstract
Biallelic ENPP1 deficiency in humans induces generalized arterial calcification of infancy (GACI) and/or autosomal recessive hypophosphatemic rickets type 2 (ARHR2). The latter is characterized by markedly increased circulating FGF23 levels and renal phosphate wasting, but aberrant skeletal manifestations associated with heterozygous ENPP1 deficiency are unknown. Here, we report three adult men with early onset osteoporosis who presented with fractures in the thoracic spine and/or left radius, mildly elevated circulating FGF23, and hypophosphatemia. Total hip bone mineral density scans demonstrated osteoporosis (Z-score < -2.5) and HRpQCT demonstrated microarchitectural defects in trabecular and cortical bone. Next-generation sequencing revealed heterozygous loss-of-function mutations in ENPP1 previously observed as biallelic mutations in infants with GACI. In addition, we present bone mass and structure data as well as plasma pyrophosphate (PPi) data of two siblings suffering from ARHR2 in comparison to their heterozygous and wild-type family members indicative of an ENPP1 gene dose effect. The skeletal phenotype in murine Enpp1 deficiency yielded nearly identical findings. Ten-week-old male Enpp1 asj/asj mice exhibited mild elevations in plasma FGF23 and hypophosphatemia, and micro-CT analysis revealed microarchitectural defects in trabecular and cortical bone of similar magnitude to HRpQCT defects observed in humans. Histomorphometry revealed mild osteomalacia and osteopenia at both 10 and 23 weeks. The biomechanical relevance of these findings was demonstrated by increased bone fragility and ductility in Enpp1 asj/asj mice. In summary, ENPP1 exerts a gene dose effect such that humans with heterozygous ENPP1 deficiency exhibit intermediate levels of plasma analytes associated with bone mineralization disturbance resulting in early onset osteoporosis. © 2019 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristin Zimmerman
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Nathan D Maulding
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Julian Stürznickel
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon von Kroge
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dillon Kavanagh
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Paul R Stabach
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Uwe Kornak
- Institute of Medical Genetics and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Steven M Tommasini
- Department of Orthoaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Mark C Horowitz
- Department of Orthoaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas O Carpenter
- Department of Orthoaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA.,Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
32
|
Corciulo C, Cronstein BN. Signaling of the Purinergic System in the Joint. Front Pharmacol 2020; 10:1591. [PMID: 32038258 PMCID: PMC6993121 DOI: 10.3389/fphar.2019.01591] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022] Open
Abstract
The joint is a complex anatomical structure consisting of different tissues, each with a particular feature, playing together to give mobility and stability at the body. All the joints have a similar composition including cartilage for reducing the friction of the movement and protecting the underlying bone, a synovial membrane that produces synovial fluid to lubricate the joint, ligaments to limit joint movement, and tendons for the interaction with muscles. Direct or indirect damage of one or more of the tissues forming the joint is the foundation of different pathological conditions. Many molecular mechanisms are involved in maintaining the joint homeostasis as well as in triggering disease development. The molecular pathway activated by the purinergic system is one of them.The purinergic signaling defines a group of receptors and intermembrane channels activated by adenosine, adenosine diphosphate, adenosine 5’-triphosphate, uridine triphosphate, and uridine diphosphate. It has been largely described as a modulator of many physiological and pathological conditions including rheumatic diseases. Here we will give an overview of the purinergic system in the joint describing its expression and function in the synovium, cartilage, ligament, tendon, and bone with a therapeutic perspective.
Collapse
Affiliation(s)
- Carmen Corciulo
- Division of Translational Medicine, Department of Medicine, NYU School of Medicine, New York, NY, United States.,Krefting Research Centre-Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Bruce N Cronstein
- Division of Translational Medicine, Department of Medicine, NYU School of Medicine, New York, NY, United States.,Division of Rheumatology, Department of Medicine, NYU School of Medicine, New York, NY, United States
| |
Collapse
|
33
|
Sato AY, Cregor M, McAndrews K, Li T, Condon KW, Plotkin LI, Bellido T. Glucocorticoid-Induced Bone Fragility Is Prevented in Female Mice by Blocking Pyk2/Anoikis Signaling. Endocrinology 2019; 160:1659-1673. [PMID: 31081900 PMCID: PMC6591015 DOI: 10.1210/en.2019-00237] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/02/2019] [Indexed: 12/16/2022]
Abstract
Excess of glucocorticoids (GCs) is a leading cause of bone fragility, and therapeutic targets are sorely needed. We report that genetic deletion or pharmacological inhibition of proline-rich tyrosine kinase 2 (Pyk2) prevents GC-induced bone loss by overriding GC effects of detachment-induced bone cell apoptosis (anoikis). In wild-type or vehicle-treated mice, GCs either prevented osteoclast apoptosis or promoted osteoblast/osteocyte apoptosis. In contrast, mice lacking Pyk2 [knockout (KO)] or treated with Pyk2 kinase inhibitor PF-431396 (PF) were protected. KO or PF-treated mice were also protected from GC-induced bone resorption, microarchitecture deterioration, and weakening of biomechanical properties. In KO and PF-treated mice, GC increased osteoclasts in bone and circulating tartrate-resistant acid phosphatase form 5b, an index of osteoclast number. However, bone surfaces covered by osteoclasts and circulating C-terminal telopeptides of type I collagen, an index of osteoclast function, were not increased. The mismatch between osteoclast number vs function induced by Pyk2 deficiency/inhibition was due to osteoclast detachment and anoikis. Further, GC prolongation of osteoclast lifespan was absent in KO and PF-treated osteoclasts, demonstrating Pyk2 as an intrinsic osteoclast-survival regulator. Circumventing Pyk2 activation preserves skeletal integrity by preventing GC effects on bone cell survival (proapoptotic for osteoblasts/osteocytes, antiapoptotic for osteoclasts) and GC-induced bone resorption. Thus, Pyk2/anoikis signaling as a therapeutic target for GC-induced osteoporosis.
Collapse
Affiliation(s)
- Amy Y Sato
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Meloney Cregor
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kevin McAndrews
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Troy Li
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Keith W Condon
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Lilian I Plotkin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Teresita Bellido
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana
- Division of Endocrinology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Center for Musculoskeletal Health, Indianapolis, Indiana
- Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, Indiana
- Correspondence: Teresita Bellido, PhD, Department of Anatomy and Cell Biology and Department of Medicine, Endocrinology, Indiana University School of Medicine, 635 Barnhill Drive, MS5045A, Indianapolis, Indiana 46202. E-mail:
| |
Collapse
|
34
|
Patel JJ, Bourne LE, Davies BK, Arnett TR, MacRae VE, Wheeler-Jones CP, Orriss IR. Differing calcification processes in cultured vascular smooth muscle cells and osteoblasts. Exp Cell Res 2019; 380:100-113. [PMID: 31004580 PMCID: PMC6520648 DOI: 10.1016/j.yexcr.2019.04.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 04/11/2019] [Accepted: 04/15/2019] [Indexed: 11/15/2022]
Abstract
Arterial medial calcification (AMC) is the deposition of calcium phosphate mineral, often as hydroxyapatite, in the medial layer of the arteries. AMC shares some similarities to skeletal mineralisation and has been associated with the transdifferentiation of vascular smooth muscle cells (VSMCs) towards an osteoblast-like phenotype. This study used primary mouse VSMCs and calvarial osteoblasts to directly compare the established and widely used in vitro models of AMC and bone formation. Significant differences were identified between osteoblasts and calcifying VSMCs. First, osteoblasts formed large mineralised bone nodules that were associated with widespread deposition of an extracellular collagenous matrix. In contrast, VSMCs formed small discrete regions of calcification that were not associated with collagen deposition and did not resemble bone. Second, calcifying VSMCs displayed a progressive reduction in cell viability over time (≤7-fold), with a 50% increase in apoptosis, whereas osteoblast and control VSMCs viability remained unchanged. Third, osteoblasts expressed high levels of alkaline phosphatase (TNAP) activity and TNAP inhibition reduced bone formation by to 90%. TNAP activity in calcifying VSMCs was ∼100-fold lower than that of bone-forming osteoblasts and cultures treated with β-glycerophosphate, a TNAP substrate, did not calcify. Furthermore, TNAP inhibition had no effect on VSMC calcification. Although, VSMC calcification was associated with increased mRNA expression of osteoblast-related genes (e.g. Runx2, osterix, osteocalcin, osteopontin), the relative expression of these genes was up to 40-fold lower in calcifying VSMCs versus bone-forming osteoblasts. In summary, calcifying VSMCs in vitro display some limited osteoblast-like characteristics but also differ in several key respects: 1) their inability to form collagen-containing bone; 2) their lack of reliance on TNAP to promote mineral deposition; and, 3) the deleterious effect of calcification on their viability.
Collapse
Affiliation(s)
- Jessal J Patel
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK; School of Life & Medical Sciences, University of Hertfordshire, Hatfield, UK
| | - Lucie E Bourne
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Bethan K Davies
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Timothy R Arnett
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Vicky E MacRae
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | | | - Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK.
| |
Collapse
|
35
|
ENPP1 in the Regulation of Mineralization and Beyond. Trends Biochem Sci 2019; 44:616-628. [PMID: 30799235 DOI: 10.1016/j.tibs.2019.01.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/26/2019] [Accepted: 01/29/2019] [Indexed: 12/24/2022]
Abstract
ENPP1 is well known for its role in regulating skeletal and soft tissue mineralization. It primarily exerts its function through the generation of pyrophosphate, a key inhibitor of hydroxyapatite formation. Several previous studies have suggested that ENPP1 also contributes to a range of human diseases including diabetes, cancer, cardiovascular disease, and osteoarthritis. In this review, we summarize the pathological roles of ENPP1 in mineralization and these soft tissue disorders. We also discuss the underlying mechanisms through which ENPP1 exerts its pathological effects. A fuller understanding of the pathways through which ENPP1 acts may help to develop novel therapeutic strategies for these commonly diagnosed morbidities.
Collapse
|
36
|
Khan T, Sinkevicius KW, Vong S, Avakian A, Leavitt MC, Malanson H, Marozsan A, Askew KL. ENPP1 enzyme replacement therapy improves blood pressure and cardiovascular function in a mouse model of generalized arterial calcification of infancy. Dis Model Mech 2018; 11:dmm.035691. [PMID: 30158213 PMCID: PMC6215426 DOI: 10.1242/dmm.035691] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/21/2018] [Indexed: 02/06/2023] Open
Abstract
Generalized arterial calcification of infancy (GACI) is a rare, life-threatening disorder caused by loss-of-function mutations in the gene encoding ectonucleotide pyrophosphatase phosphodiesterase 1 (ENPP1), which normally hydrolyzes extracellular ATP into AMP and pyrophosphate (PPi). The disease is characterized by extensive arterial calcification and stenosis of large- and medium-sized vessels, leading to vascular-related complications of hypertension and heart failure. There is currently no effective treatment available, but bisphosphonates – nonhydrolyzable PPi analogs – are being used off-label to reduce arterial calcification, although this has no reported impact on the hypertension and cardiac dysfunction features of GACI. In this study, the efficacy of a recombinant human ENPP1 protein therapeutic (rhENPP1) was tested in Enpp1asj-2J homozygous mice (Asj-2J or Asj-2J hom), a model previously described to show extensive mineralization in the arterial vasculature, similar to GACI patients. In a disease prevention study, Asj-2J mice treated with rhENPP1 for 3 weeks showed >95% reduction in aorta calcification. Terminal hemodynamics and echocardiography imaging of Asj-2J mice also revealed that a 6-week rhENPP1 treatment normalized elevated arterial and left ventricular pressure, which translated into significant improvements in myocardial compliance, contractility, heart workload and global cardiovascular efficiency. This study suggests that ENPP1 enzyme replacement therapy could be a more effective GACI therapeutic than bisphosphonates, treating not just the vascular calcification, but also the hypertension that eventually leads to cardiac failure in GACI patients. Summary: ENPP1 enzyme replacement therapy can have important implications for generalized arterial calcification of infancy by treating both vascular calcification and hypertension, which are the leading causes of cardiac failure and mortality in patients.
Collapse
Affiliation(s)
- Tayeba Khan
- Alexion Pharmaceuticals, Lexington, MA 02421, USA
| | | | - Sylvia Vong
- Alexion Pharmaceuticals, New Haven, CT 06510, USA
| | | | | | | | | | - Kim L Askew
- Alexion Pharmaceuticals, Lexington, MA 02421, USA
| |
Collapse
|
37
|
Jin Y, Cong Q, Gvozdenovic-Jeremic J, Hu J, Zhang Y, Terkeltaub R, Yang Y. Enpp1 inhibits ectopic joint calcification and maintains articular chondrocytes by repressing hedgehog signaling. Development 2018; 145:dev.164830. [PMID: 30111653 DOI: 10.1242/dev.164830] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/12/2018] [Indexed: 01/15/2023]
Abstract
The differentiated phenotype of articular chondrocytes of synovial joints needs to be maintained throughout life. Disruption of the articular cartilage, frequently associated with chondrocyte hypertrophy and calcification, is a central feature in osteoarthritis (OA). However, the molecular mechanisms whereby phenotypes of articular chondrocytes are maintained and pathological calcification is inhibited remain poorly understood. Recently, the ecto-enzyme Enpp1, a suppressor of pathological calcification, was reported to be decreased in joint cartilage with OA in both human and mouse, and Enpp1 deficiency causes joint calcification. Here, we found that hedgehog (Hh) signaling activation contributes to ectopic joint calcification in the Enpp1-/- mice. In the Enpp1-/- joints, Hh signaling was upregulated. Further activation of Hh signaling by removing the patched 1 gene in the Enpp1-/- mice enhanced ectopic joint calcification, whereas removing Gli2 partially rescued the ectopic calcification phenotype. In addition, reduction of Gαs in the Enpp1-/- mice enhanced joint calcification, suggesting that Enpp1 inhibits Hh signaling and chondrocyte hypertrophy by activating Gαs-PKA signaling. Our findings provide new insights into the mechanisms underlying Enpp1 regulation of joint integrity.
Collapse
Affiliation(s)
- Yunyun Jin
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, MA 02115, USA.,Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Qian Cong
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, MA 02115, USA
| | | | - Jiajie Hu
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Yiqun Zhang
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Robert Terkeltaub
- Department of Medicine, Veterans Affairs Healthcare System, University of California San Diego, 111K, 3350 La Jolla Village Dr., San Diego, CA 92161, USA
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, MA 02115, USA
| |
Collapse
|
38
|
Tsourdi E, Jähn K, Rauner M, Busse B, Bonewald LF. Physiological and pathological osteocytic osteolysis. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2018; 18:292-303. [PMID: 30179206 PMCID: PMC6146198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Osteocytes, the most abundant bone cell in the adult skeleton, can function as mechanosensors directing osteoblast and osteoclast function in order to maintain optimal load bearing bone in addition to functioning as endocrine cells regulating phosphate metabolism. A controversial function, previously overlooked or denied, has been osteocytes as regulators of calcium metabolism. Early histologists upon observing enlarged osteocyte lacunae in bone sections proposed that mature osteocytes could remove their perilacunar matrix, a term called "osteocytic osteolysis". New insights into this process have occurred during the last decade using novel technology thereby providing a means to identify molecular mechanisms responsible for osteocytic osteolysis. As release of calcium from a mineralized matrix requires a more acidic pH and specialized enzymes, it was proposed that osteocytes may utilize similar molecular mechanisms as osteoclasts to remove mineral. The idea that a cell descended from mesenchymal progenitors (the osteocyte) could function similarly to a cell descended from hematopoietic progenitors (the osteoclast) was challenged as being improbable. Here we review the molecular mechanisms behind this osteocyte function, the role of osteocytic osteolysis in health and disease, and the capacity of the osteocyte to reverse the osteolytic process by replacing the removed matrix, a revived osteoblast function.
Collapse
Affiliation(s)
- Elena Tsourdi
- Department of Medicine III, Technische Universität Dresden Medical Center, Dresden, Germany,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany,Corresponding author: Elena Tsourdi, M.D., Division of Endocrinology, Diabetes and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden Medical Center, Fetscherstrasse 74, 01307 Dresden, Germany E-mail:
| | - Katharina Jähn
- Department for Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martina Rauner
- Department of Medicine III, Technische Universität Dresden Medical Center, Dresden, Germany,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Björn Busse
- Department for Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lynda F. Bonewald
- Indiana Center for Musculoskeletal Health, VanNuys Medical Science Building, Indianapolis, USA
| |
Collapse
|
39
|
Wolf M, Ao M, Chavez M, Kolli T, Thumbigere-Math V, Becker K, Chu E, Jäger A, Somerman M, Foster B. Reduced Orthodontic Tooth Movement in Enpp1 Mutant Mice with Hypercementosis. J Dent Res 2018; 97:937-945. [PMID: 29533727 PMCID: PMC6728553 DOI: 10.1177/0022034518759295] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Previous studies revealed that cementum formation is tightly regulated by inorganic pyrophosphate (PPi), a mineralization inhibitor. Local PPi concentrations are determined by regulators, including ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1), which increases PPi concentrations by adenosine triphosphate hydrolysis. Orthodontic forces stimulate alveolar bone remodelling, leading to orthodontic tooth movement (OTM). To better understand how disturbed mineral metabolism and the resulting altered periodontal structures affect OTM, we employed Enpp1 mutant mice that feature reduced PPi and increased cervical cementum in a model of OTM induced by a stretched closed-coil spring ligated between the maxillary left first molar and maxillary incisors. We analyzed tooth movement, osteoclast/odontoclast response, and tooth root resorption by micro-computed tomography, histology, histomorphometry, and immunohistochemistry. Preoperatively, we noted an altered periodontium in Enpp1 mutant mice, with significantly increased periodontal ligament (PDL) volume and thickness, as well as increased PDL-bone/tooth root surface area, compared to wild-type (WT) controls. After 11 d of orthodontic treatment, Enpp1 mutant mice displayed 38% reduced tooth movement versus WT mice. Molar roots in Enpp1 mutant mice exhibited less change in PDL width in compression and tension zones compared to WT mice. Root resorption was noted in both groups with no difference in average depths, but resorption lacunae in Enpp1 mutant mice were almost entirely limited to cementum, with 150% increased cementum resorption and 92% decreased dentin resorption. Osteoclast/odontoclast cells were reduced by 64% in Enpp1 mutant mice, with a predominance of tartrate-resistant acid phosphatase (TRAP)-positive cells on root surfaces, compared to WT mice. Increased numbers of TRAP-positive cells on root surfaces were associated with robust immunolocalization of osteopontin (OPN) and receptor-activator of NF-κB ligand (RANKL). Collectively, reduced response to orthodontic forces, decreased tooth movement, and altered osteoclast/odontoclast distribution suggests Enpp1 loss of function has direct effects on clastic function/recruitment and/or indirect effects on periodontal remodeling via altered periodontal structure or tissue mineralization.
Collapse
Affiliation(s)
- M. Wolf
- Department of Orthodontics, Aachen
University, Aachen, Germany
- National Institute of Arthritis and
Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH),
Bethesda, MD, USA
| | - M. Ao
- National Institute of Arthritis and
Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH),
Bethesda, MD, USA
| | - M.B. Chavez
- Division of Biosciences, College of
Dentistry, The Ohio State University, Columbus, OH, USA
| | - T.N. Kolli
- Division of Biosciences, College of
Dentistry, The Ohio State University, Columbus, OH, USA
| | - V. Thumbigere-Math
- National Institute of Arthritis and
Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH),
Bethesda, MD, USA
- Division of Periodontics, School of
Dentistry, University of Maryland, Baltimore, MD, USA
| | - K. Becker
- Department of Orthodontics,
Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - E.Y. Chu
- National Institute of Arthritis and
Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH),
Bethesda, MD, USA
| | - A. Jäger
- Department of Orthodontics, University
of Bonn, Bonn, Germany
| | - M.J. Somerman
- National Institute of Arthritis and
Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH),
Bethesda, MD, USA
| | - B.L. Foster
- Division of Biosciences, College of
Dentistry, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
40
|
Patel JJ, Zhu D, Opdebeeck B, D’Haese P, Millán JL, Bourne LE, Wheeler-Jones CPD, Arnett TR, MacRae VE, Orriss IR. Inhibition of arterial medial calcification and bone mineralization by extracellular nucleotides: The same functional effect mediated by different cellular mechanisms. J Cell Physiol 2018; 233:3230-3243. [PMID: 28976001 PMCID: PMC5792173 DOI: 10.1002/jcp.26166] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/22/2017] [Indexed: 12/30/2022]
Abstract
Arterial medial calcification (AMC) is thought to share some outward similarities to skeletal mineralization and has been associated with the transdifferentiation of vascular smooth muscle cells (VSMCs) to an osteoblast-like phenotype. ATP and UTP have previously been shown to inhibit bone mineralization. This investigation compared the effects of extracellular nucleotides on calcification in VSMCs with those seen in osteoblasts. ATP, UTP and the ubiquitous mineralization inhibitor, pyrophosphate (PPi ), dose dependently inhibited VSMC calcification by ≤85%. Culture of VSMCs in calcifying conditions was associated with an increase in apoptosis; treatment with ATP, UTP, and PPi reduced apoptosis to levels seen in non-calcifying cells. Extracellular nucleotides had no effect on osteoblast viability. Basal alkaline phosphatase (TNAP) activity was over 100-fold higher in osteoblasts than VSMCs. ATP and UTP reduced osteoblast TNAP activity (≤50%) but stimulated VSMC TNAP activity (≤88%). The effects of extracellular nucleotides on VSMC calcification, cell viability and TNAP activity were unchanged by deletion or inhibition of the P2Y2 receptor. Conversely, the actions of ATP/UTP on bone mineralization and TNAP activity were attenuated in osteoblasts lacking the P2Y2 receptor. Ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (NPP1) hydrolyses ATP and UTP to produce PPi . In both VSMCs and osteoblasts, deletion of NPP1 blunted the inhibitory effects of extracellular nucleotides suggesting involvement of P2 receptor independent pathways. Our results show that although the overall functional effect of extracellular nucleotides on AMC and bone mineralization is similar there are clear differences in the cellular mechanisms mediating these actions.
Collapse
Affiliation(s)
- JJ Patel
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - D Zhu
- Guangzhou Institute of Cardiovascular Disease, School of Basic Medical Sciences, Guangzhou Medical University, China
| | - B Opdebeeck
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium
| | - P D’Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium
| | - JL Millán
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - LE Bourne
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - CPD Wheeler-Jones
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - TR Arnett
- Department of Cell and Developmental Biology, University College London, London, UK
| | - VE MacRae
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - IR Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| |
Collapse
|
41
|
ATP-degrading ENPP1 is required for survival (or persistence) of long-lived plasma cells. Sci Rep 2017; 7:17867. [PMID: 29259245 PMCID: PMC5736562 DOI: 10.1038/s41598-017-18028-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/05/2017] [Indexed: 02/05/2023] Open
Abstract
Survival of antibody-secreting plasma cells (PCs) is vital for sustained antibody production. However, it remains poorly understood how long-lived PCs (LLPCs) are generated and maintained. Here we report that ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) is preferentially upregulated in bone marrow LLPCs compared with their splenic short-lived counterparts (SLPCs). We studied ENPP1-deficient mice (Enpp1−/−) to determine how the enzyme affects PC biology. Although Enpp1−/− mice generated normal levels of germinal center B cells and plasmablasts in periphery, they produced significantly reduced numbers of LLPCs following immunization with T-dependent antigens or infection with plasmodium C. chabaudi. Bone marrow chimeric mice showed B cell intrinsic effect of ENPP1 selectively on generation of bone marrow as well as splenic LLPCs. Moreover, Enpp1−/− PCs took up less glucose and had lower levels of glycolysis than those of wild-type controls. Thus, ENPP1 deficiency confers an energetic disadvantage to PCs for long-term survival and antibody production.
Collapse
|
42
|
Thumbigere-Math V, Alqadi A, Chalmers NI, Chavez MB, Chu EY, Collins MT, Ferreira CR, FitzGerald K, Gafni RI, Gahl WA, Hsu KS, Ramnitz MS, Somerman MJ, Ziegler SG, Foster BL. Hypercementosis Associated with ENPP1 Mutations and GACI. J Dent Res 2017; 97:432-441. [PMID: 29244957 DOI: 10.1177/0022034517744773] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mineralization of bones and teeth is tightly regulated by levels of extracellular inorganic phosphate (Pi) and pyrophosphate (PPi). Three regulators that control pericellular concentrations of Pi and PPi include tissue-nonspecific alkaline phosphatase (TNAP), progressive ankylosis protein (ANK), and ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1). Inactivation of these factors results in mineralization disorders affecting teeth and their supporting structures. This study for the first time analyzed the effect of decreased PPi on dental development in individuals with generalized arterial calcification of infancy (GACI) due to loss-of-function mutations in the ENPP1 gene. Four of the 5 subjects reported a history of infraocclusion, overretained primary teeth, ankylosis, and/or slow orthodontic tooth movement, suggesting altered mineral metabolism contributing to disrupted tooth movement and exfoliation. All subjects had radiographic evidence of unusually protruding cervical root morphology in primary and/or secondary dentitions. High-resolution micro-computed tomography (micro-CT) analyses of extracted primary teeth from 3 GACI subjects revealed 4-fold increased cervical cementum thickness ( P = 0.00007) and a 23% increase in cementum density ( P = 0.009) compared to age-matched healthy control teeth. There were no differences in enamel and dentin densities between GACI and control teeth. Histology revealed dramatically expanded cervical cementum in GACI teeth, including cementocyte-like cells and unusual patterns of cementum resorption and repair. Micro-CT analysis of Enpp1 mutant mouse molars revealed 4-fold increased acellular cementum thickness ( P = 0.002) and 5-fold increased cementum volume ( P = 0.002), with no changes in enamel or dentin. Immunohistochemistry identified elevated ENPP1 expression in cementoblasts of human and mouse control teeth. Collectively, these findings reveal a novel dental phenotype in GACI and identify ENPP1 genetic mutations associated with hypercementosis. The sensitivity of cementum to reduced PPi levels in both human and mouse teeth establishes this as a well-conserved and fundamental biological process directing cementogenesis across species (ClinicalTrials.gov NCT00369421).
Collapse
Affiliation(s)
- V Thumbigere-Math
- 1 National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA.,2 Division of Periodontics, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - A Alqadi
- 3 Division of Public and Child Dental Health, Dublin Dental University Hospital, Dublin, Ireland
| | - N I Chalmers
- 4 Analytics and Publication, DentaQuest Institute, Westborough, MA, USA
| | - M B Chavez
- 5 Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - E Y Chu
- 1 National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - M T Collins
- 6 Section on Skeletal Disorders and Mineral Homeostasis, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - C R Ferreira
- 7 National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Bethesda, MD, USA.,8 Division of Genetics and Metabolism, Children's National Health System, Washington, DC, USA
| | - K FitzGerald
- 3 Division of Public and Child Dental Health, Dublin Dental University Hospital, Dublin, Ireland.,9 Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| | - R I Gafni
- 6 Section on Skeletal Disorders and Mineral Homeostasis, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - W A Gahl
- 7 National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - K S Hsu
- 7 National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - M S Ramnitz
- 6 Section on Skeletal Disorders and Mineral Homeostasis, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - M J Somerman
- 1 National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - S G Ziegler
- 10 Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - B L Foster
- 5 Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
43
|
Orriss IR, Guneri D, Hajjawi MOR, Shaw K, Patel JJ, Arnett TR. Activation of the P2Y 2 receptor regulates bone cell function by enhancing ATP release. J Endocrinol 2017; 233:341-356. [PMID: 28420708 DOI: 10.1530/joe-17-0042] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 04/18/2017] [Indexed: 02/02/2023]
Abstract
Bone cells constitutively release ATP into the extracellular environment where it acts locally via P2 receptors to regulate bone cell function. Whilst P2Y2 receptor stimulation regulates bone mineralisation, the functional effects of this receptor in osteoclasts remain unknown. This investigation used the P2Y2 receptor knockout (P2Y2R-/- ) mouse model to investigate the role of this receptor in bone. MicroCT analysis of P2Y2R-/- mice demonstrated age-related increases in trabecular bone volume (≤48%), number (≤30%) and thickness (≤17%). In vitro P2Y2R-/- osteoblasts displayed a 3-fold increase in bone formation and alkaline phosphatase activity, whilst P2Y2R-/- osteoclasts exhibited a 65% reduction in resorptive activity. Serum cross-linked C-telopeptide levels (CTX, resorption marker) were also decreased (≤35%). The resorption defect in P2Y2R-/- osteoclasts was rescued by the addition of exogenous ATP, suggesting that an ATP deficit could be a key factor in the reduced function of these cells. In agreement, we found that basal ATP release was reduced up to 53% in P2Y2R-/- osteoclasts. The P2Y2 receptor agonists, UTP and 2-thioUTP, increased osteoclast activity and ATP release in wild-type but not in P2Y2R-/- cells. This indicates that the P2Y2 receptor may regulate osteoclast function indirectly by promoting ATP release. UTP and 2-thioUTP also stimulate ATP release from osteoblasts suggesting that the P2Y2 receptor exerts a similar function in these cells. Taken together, our findings are consistent with the notion that the primary action of P2Y2 receptor signalling in bone is to regulate extracellular ATP levels.
Collapse
Affiliation(s)
- Isabel R Orriss
- Department of Comparative Biomedical SciencesRoyal Veterinary College, London, UK
| | - Dilek Guneri
- Department of Comparative Biomedical SciencesRoyal Veterinary College, London, UK
| | - Mark O R Hajjawi
- Department of Cell & Developmental BiologyUniversity College London, London, UK
| | - Kristy Shaw
- Department of Comparative Biomedical SciencesRoyal Veterinary College, London, UK
| | - Jessal J Patel
- Department of Comparative Biomedical SciencesRoyal Veterinary College, London, UK
| | - Timothy R Arnett
- Department of Cell & Developmental BiologyUniversity College London, London, UK
| |
Collapse
|
44
|
Constant M, Nicot R, Vieira AR, Raoul G, Sciote JJ, Ferri J. Condylar geometry variation is associated with ENPP1 variant in a population of patients with dento-facial deformities. J Craniomaxillofac Surg 2017; 45:826-830. [PMID: 28381371 DOI: 10.1016/j.jcms.2017.02.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 01/24/2017] [Accepted: 02/17/2017] [Indexed: 01/08/2023] Open
Abstract
PURPOSE Bone remodeling is essential in maintaining bone health. Considering that ENPP1 contributes to bone geometry and bone mineralization, the aim of our study was to analyze the association between single-nucleotide polymorphisms (SNPs) of ENPP1 and condylar remodeling. MATERIALS AND METHODS A total of 156 patients undergoing orthodontic and maxillofacial surgery treatment for correction of malocclusion were included in this prospective study. Saliva samples from all subjects were used for DNA extraction and genotyping. Four ENPP1 SNPs were selected and tested to determine whether specific allelic variants are correlated with condylar remodeling. The criteria of condylar remodeling chosen were the ratio between each side of condylar height or surface differences on a dental panoramic of each patient. A diagnostic threshold was set at 15% difference between both sides. RESULTS The ENPP1 SNP rs9373000 showed a statistically significant association with condylar height ratio >15% (p = 0.012). The GG genotype was found to be a protective factor against condylar height decrease (p = 0.003). CONCLUSION This study identifies the genetic variant rs9373000 as a potentially causal variant for mandibular condyle geometry variation for patients presenting with dento-facial deformities.
Collapse
Affiliation(s)
- Marion Constant
- Univ. Lille, Oral and Maxillofacial Department, Roger Salengro Hospital, CHU Lille, F-59000 Lille, France.
| | - Romain Nicot
- Univ. Lille, Oral and Maxillofacial Department, Roger Salengro Hospital, CHU Lille, F-59000 Lille, France.
| | - Alexandre R Vieira
- Department of Oral Biology, University of Pittsburgh School of Dental Medicine, 3501 Terrace St, Pittsburgh PA 15261, USA.
| | - Gwenael Raoul
- Univ. Lille, Oral and Maxillofacial Department, Roger Salengro Hospital, CHU Lille, INSERM U 1008, Controlled Drug Delivery Systems and Biomaterials, F-59000 Lille, France.
| | | | - Joel Ferri
- Univ. Lille, Oral and Maxillofacial Department, Roger Salengro Hospital, CHU Lille, INSERM U 1008, Controlled Drug Delivery Systems and Biomaterials, F-59000 Lille, France.
| |
Collapse
|
45
|
New perspectives on rare connective tissue calcifying diseases. Curr Opin Pharmacol 2016; 28:14-23. [DOI: 10.1016/j.coph.2016.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 01/27/2016] [Accepted: 02/08/2016] [Indexed: 12/27/2022]
|
46
|
Prideaux M, Findlay DM, Atkins GJ. Osteocytes: The master cells in bone remodelling. Curr Opin Pharmacol 2016; 28:24-30. [DOI: 10.1016/j.coph.2016.02.003] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 01/14/2016] [Accepted: 02/09/2016] [Indexed: 10/22/2022]
|
47
|
Li Q, Kingman J, Sundberg JP, Levine MA, Uitto J. Dual Effects of Bisphosphonates on Ectopic Skin and Vascular Soft Tissue Mineralization versus Bone Microarchitecture in a Mouse Model of Generalized Arterial Calcification of Infancy. J Invest Dermatol 2016; 136:275-283. [PMID: 26763447 PMCID: PMC4731049 DOI: 10.1038/jid.2015.377] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/02/2015] [Accepted: 09/11/2015] [Indexed: 12/30/2022]
Abstract
Generalized arterial calcification of infancy (GACI) is an intractable ectopic mineralization disorder caused by mutations in the ENPP1 gene resulting in reduced plasma inorganic pyrophosphate levels. We previously characterized the Enpp1asj mutant mouse as a model of GACI, and we have now explored the potential efficacy of bisphosphonates, non-hydrolyzable PPi analogs, in preventing ectopic mineralization in these mice. These mice were maintained on either basic diet (control) or diets containing etidronate or alendronate in three different concentrations (experimental). Considering low bioavailability of bisphosphonates when administered orally, subsequent studies tested the mice with subcutaneous injections of etidronate. The treatments were initiated at 4 weeks of age, and the degree of mineralization was assessed at 12 weeks of age by quantitation of calcium deposits in the muzzle skin containing dermal sheath of vibrissae and in aorta. We found that bisphosphonate treatments significantly reduced mineralization in skin and aorta. These changes in treated mice were accompanied with restoration of their bone microarchitecture, determined bymicrocomputed tomography. The inhibitory capacity of bisphosphonates, with mechanistic implications, was confirmed in a cell-based mineralization assay in vitro. Collectively, these results suggest that bisphosphonate treatment may be beneficial by a dual effect for preventing ectopic soft tissue mineralization while correcting decreased bone mineralization in GACI caused by ENPP1 mutations.
Collapse
Affiliation(s)
- Qiaoli Li
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| | - Joshua Kingman
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Michael A Levine
- Division of Endocrinology, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Jouni Uitto
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
48
|
Orriss IR, Arnett TR, Russell RGG. Pyrophosphate: a key inhibitor of mineralisation. Curr Opin Pharmacol 2016; 28:57-68. [PMID: 27061894 DOI: 10.1016/j.coph.2016.03.003] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 03/18/2016] [Accepted: 03/24/2016] [Indexed: 12/20/2022]
Abstract
Inorganic pyrophosphate has long been known as a by-product of many intracellular biosynthetic reactions, and was first identified as a key endogenous inhibitor of biomineralisation in the 1960s. The major source of pyrophosphate appears to be extracellular ATP, which is released from cells in a controlled manner. Once released, ATP can be rapidly hydrolysed by ecto-nucleotide pyrophosphatase/phosphodiesterases to produce pyrophosphate. The main action of pyrophosphate is to directly inhibit hydroxyapatite formation thereby acting as a physiological 'water-softener'. Evidence suggests pyrophosphate may also act as a signalling molecule to influence gene expression and regulate its own production and breakdown. This review will summarise our current understanding of pyrophosphate metabolism and how it regulates bone mineralisation and prevents harmful soft tissue calcification.
Collapse
Affiliation(s)
- Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK.
| | - Timothy R Arnett
- Department of Cell and Developmental Biology, University College London, London, UK
| | - R Graham G Russell
- The Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford, UK; The Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
| |
Collapse
|
49
|
Orriss IR, Key ML, Hajjawi MOR, Millán JL, Arnett TR. Acidosis is a key regulator of osteoblast ecto-nucleotidase pyrophosphatase/phosphodiesterase 1 (NPP1) expression and activity. J Cell Physiol 2015; 230:3049-56. [PMID: 26033523 PMCID: PMC4549203 DOI: 10.1002/jcp.25041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/07/2015] [Indexed: 01/19/2023]
Abstract
Previous work has shown that acidosis prevents bone nodule formation by osteoblasts in vitro by inhibiting mineralisation of the collagenous matrix. The ratio of phosphate (Pi) to pyrophosphate (PPi) in the bone microenvironment is a fundamental regulator of bone mineralisation. Both Pi and PPi, a potent inhibitor of mineralisation, are generated from extracellular nucleotides by the actions of ecto‐nucleotidases. This study investigated the expression and activity of ecto‐nucleotidases by osteoblasts under normal and acid conditions. We found that osteoblasts express mRNA for a number of ecto‐nucleotidases including NTPdase 1–6 (ecto‐nucleoside triphosphate diphosphohydrolase) and NPP1‐3 (ecto‐nucleotide pyrophosphatase/phosphodiesterase). The rank order of mRNA expression in differentiating rat osteoblasts (day 7) was Enpp1 > NTPdase 4 > NTPdase 6 > NTPdase 5 > alkaline phosphatase > ecto‐5‐nucleotidase > Enpp3 > NTPdase 1 > NTPdase 3 > Enpp2 > NTPdase 2. Acidosis (pH 6.9) upregulated NPP1 mRNA (2.8‐fold) and protein expression at all stages of osteoblast differentiation compared to physiological pH (pH 7.4); expression of other ecto‐nucleotidases was unaffected. Furthermore, total NPP activity was increased up to 53% in osteoblasts cultured in acid conditions (P < 0.001). Release of ATP, one of the key substrates for NPP1, from osteoblasts, was unaffected by acidosis. Further studies showed that mineralised bone formation by osteoblasts cultured from NPP1 knockout mice was increased compared with wildtypes (2.5‐fold, P < 0.001) and was partially resistant to the inhibitory effect of acidosis. These results indicate that increased NPP1 expression and activity might contribute to the decreased mineralisation observed when osteoblasts are exposed to acid conditions. J. Cell. Physiol. 230: 3049–3056, 2015. © 2015 The Authors. Journal of Cellular Physiology Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK.,Department of Cell and Developmental Biology, University College London, London, UK
| | - Michelle L Key
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Mark O R Hajjawi
- Department of Cell and Developmental Biology, University College London, London, UK
| | - José L Millán
- Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Timothy R Arnett
- Department of Cell and Developmental Biology, University College London, London, UK
| |
Collapse
|
50
|
Huesa C, Staines KA, Millán JL, MacRae VE. Effects of etidronate on the Enpp1⁻/⁻ mouse model of generalized arterial calcification of infancy. Int J Mol Med 2015; 36:159-65. [PMID: 25975272 PMCID: PMC4494596 DOI: 10.3892/ijmm.2015.2212] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/22/2015] [Indexed: 12/23/2022] Open
Abstract
Generalized arterial calcification of infancy (GACI) is an autosomal recessive disorder of spontaneous infantile arterial and periarticular calcification which is attributed to mutations in the ectonucleotide pyrophosphatase/phosphodiesterase 1 (Enpp1) gene. Whilst the bisphosphonate, etidronate, is currently used off-label for the treatment for GACI, recent studies have highlighted its detrimental effects on bone mineralisation. In the present study, we used the Enpp1-/- mouse model of GACI to examine the effects of etidronate treatment (100 µg/kg), on vascular and skeletal calcification. Micro-computed tomography (µCT) analysis revealed a significant decrease in trabecular bone mass, as reflected by the decrease in trabecular bone volume/tissue volume (BV/TV; %), trabecular thickness, trabecular separation, trabecular number and pattern factor (P<0.05) in the Enpp1-/- mice in comparison to the wild-type (WT) mice. Mechanical testing revealed that in the WT mice, treatment with etidronate significantly improved work to fracture and increased work post-failure (P<0.05, in comparison to the vehicle-treated WT mice). This significant increase, however, was not observed in the Enpp1-/- mice. Treatment with etidronate had no effect on bone parameters in the WT mice; however, the Enpp1-/- mice displayed an increased structural model index (SMI; P<0.05). We used a recently developed 3D µCT protocol to reconstruct and quantify the extensive aortic calcification in Enpp1-/- mice in comparison to the WT mice. However, treatment with etidronate did not prevent de novo calcification, and did not arrest the progression of established calcification of the aorta.
Collapse
Affiliation(s)
- Carmen Huesa
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| | | | - Jose Luis Millán
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Vicky E MacRae
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|