1
|
Deering J, Buss DJ, Kröger R, Vali H, Lagos MJ, Reznikov N, McKee MD. Bone mineralization and the effects of elevated osteopontin: from symmetry-breaking foci to 3D space-filling tessellation. Faraday Discuss 2025. [PMID: 40439195 PMCID: PMC12123585 DOI: 10.1039/d5fd00013k] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/19/2025] [Indexed: 06/02/2025]
Abstract
At the nanoscale, lamellar bone tissue mineralization ensues via heteronucleation of small mineral foci within the osteoid. The foci grow to produce a mature, volume-filling tessellation pattern at the micrometer-scale. Mineralization-inhibiting osteopontin (OPN) mediates this bone mineralization pathway and, eventually, the microscale properties of bone tissue. Using 2D and 3D electron microscopy, here we have assessed how the abundance of OPN can affect nanoscale mineralization, mineral ripening, and microscale patterning of mineral in normal wild-type mouse bone, and we compare that to mutant mouse models having elevated OPN (Fgf23-/- and Hyp mice). When OPN is elevated, volume-filling mineral tessellation was incomplete (showing a four-fold increase in mineral surface area in the vicinity of the mineralization front in Hyp bone). Immunogold labeling showed excessive OPN in the foci, suggesting an arrest of their growth and an interruption of the pathway towards microscale tessellation. In Fgf23-/- mice, electron tomography and 3D focused ion beam-scanning electron microscopy (FIB-SEM) imaging of mineral foci show instances of core-shell morphology with crystalline mineral confined to the focus interior, and an amorphous nanogranular texture persisting in the outer shell. Electron energy-loss spectroscopy, which is sensitive to nanoscale elemental composition, showed a lower Ca/P ratio at the periphery of Hyp foci, consistent with a more amorphous mineral character, suggesting that OPN may play a role in delaying the amorphous-to-crystalline transition. These aspects of nanoscale mineral maturation in mutant mice having elevated OPN implicate this protein as a fine-tuning regulator of mineralization kinetics, mineral composition, and mechanical properties of bone.
Collapse
Affiliation(s)
- Joseph Deering
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Strathcona Anatomy and Dentistry Bldg, Rm M73 3640 University Street, Montreal, QC, H3A 0C7, Canada.
| | - Daniel J Buss
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, H3A 0C7, Canada
| | - Roland Kröger
- Department of Physics, University of York, York, YO10 5DD, UK
| | - Hojatollah Vali
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, H3A 0C7, Canada
| | - Maureen J Lagos
- Department of Materials Science and Engineering, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Natalie Reznikov
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Strathcona Anatomy and Dentistry Bldg, Rm M73 3640 University Street, Montreal, QC, H3A 0C7, Canada.
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, H3A 0C7, Canada
- Department of Bioengineering, Faculty of Engineering, McGill University, Montreal, QC, H3A 0E9, Canada
| | - Marc D McKee
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Strathcona Anatomy and Dentistry Bldg, Rm M73 3640 University Street, Montreal, QC, H3A 0C7, Canada.
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, H3A 0C7, Canada
| |
Collapse
|
2
|
Mardiyantoro F, Chiba N, Seong CH, Tada R, Ohnishi T, Nakamura N, Matsuguchi T. Two-sided function of osteopontin during osteoblast differentiation. J Biochem 2025; 177:121-131. [PMID: 39658208 DOI: 10.1093/jb/mvae080] [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: 08/22/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024] Open
Abstract
Osteopontin (OPN) is expressed in various cell types including osteoblasts. OPN expression level is robustly increased during osteoblast differentiation. Although OPN was initially found as a secretory protein (sOPN), recent reports identified the intracellular isoform of OPN (iOPN). Distinct functions of each OPN isoform in osteoblasts, however, are not well established. Here, using the Tet-On inducible expression system, we examined the role of each OPN isoform during osteoblast differentiation. Induced overexpression of wild type OPN (wtOPN), which includes both sOPN and iOPN, significantly increased matrix mineralization and osteogenic marker gene expression during osteogenic differentiation induced by either ascorbic acid or bone morphogenetic protein (BMP) 9. In contrast, these osteogenic differentiation processes were significantly inhibited by the specific overexpression of iOPN. Furthermore, the addition of recombinant OPN or neutralizing anti-OPN antibody to the culture medium exerted promotive or inhibitory effect on osteoblast differentiation, respectively. These data strongly indicate that iOPN exerts inhibitory effects on osteoblast differentiation, whereas sOPN exerts positive effects. We also found that the secretion process of OPN is positively regulated by c-Jun N-terminal kinase (JNK) activity in osteoblasts.
Collapse
Affiliation(s)
- Fredy Mardiyantoro
- Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
- Department of Oral Biochemistry, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
- Department of Oral and Maxillofacial Surgery, Dentistry Faculty of Brawijaya University, Jalan Veteran 65145, Malang, Indonesia
| | - Norika Chiba
- Department of Oral Biochemistry, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Chang-Hwan Seong
- Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
- Department of Oral Biochemistry, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
- Department of Biosignals and Inheritance, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Ryohei Tada
- Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
- Department of Oral Biochemistry, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Tomokazu Ohnishi
- Department of Oral Biochemistry, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Norifumi Nakamura
- Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Tetsuya Matsuguchi
- Department of Oral Biochemistry, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| |
Collapse
|
3
|
Buss DJ, Deering J, Reznikov N, McKee MD. Understanding the structural biology of osteomalacia through multiscale 3D X-ray and electron tomographic imaging: a review of X-linked hypophosphatemia, the Hyp mouse model, and imaging methods. JBMR Plus 2025; 9:ziae176. [PMID: 39896117 PMCID: PMC11783288 DOI: 10.1093/jbmrpl/ziae176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/10/2024] [Accepted: 12/28/2024] [Indexed: 02/04/2025] Open
Abstract
Biomineralization in bones and teeth is a highly regulated extracellular event. In the skeleton, mineralization at the tissue level is controlled within the collagenous extracellular matrix by both circulating and local factors. While systemic regulation of mineral ion homeostasis has been well-studied over many decades, much less is known about the regulation of mineralization at the local level directly within the extracellular matrix. Some local regulators have been identified, such as tissue-nonspecific alkaline phosphatase (TNAP), phosphate-regulating endopeptidase homolog X-linked (PHEX), pyrophosphate, and osteopontin, and others are currently under investigation. Dysregulation of the actions of enzyme-inhibitor substrate pairs engaged in mineralization (as we describe by the Stenciling Principle for extracellular matrix mineralization) leads to osteomalacic "soft bone" diseases, such as hypophosphatasia (HPP) and X-linked hypophosphatemia (XLH). This review addresses how advances in 3D imaging tools and software now allow contextual and correlative viewing and interpretation of mineralized tissue structure across most length scales. Contextualized and integrated 3D multiscale data obtained from these imaging modalities have afforded an unprecedented structural biology view of bone from the macroscale to the nanoscale. Such correlated volume imaging data is highly quantitative, providing not only an integrated view of the skeleton in health, but also a means to observe alterations that occur in disease. In the context of the many hierarchical levels of skeletal organization, here we summarize structural features of bone over multiple length scales, with a focus on nano- and microscale features as viewed by X-ray and electron tomography imaging methods (submicron μCT and FIB-SEM). We additionally summarize structural changes observed after dysregulation of the mineralization pathway, focusing here on the Hyp mouse model for XLH. More specifically, we summarize how mineral patterns/packs at the microscale (3D crossfibrillar mineral tessellation), and how this is defective in Hyp mouse bone and Hyp enthesis fibrocartilage.
Collapse
Affiliation(s)
- Daniel J Buss
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, School of Biomedical Sciences, McGill University, Montreal, QC H3A 0C7, Canada
| | - Joseph Deering
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
| | - Natalie Reznikov
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, School of Biomedical Sciences, McGill University, Montreal, QC H3A 0C7, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
- Department of Bioengineering, Faculty of Engineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Marc D McKee
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, School of Biomedical Sciences, McGill University, Montreal, QC H3A 0C7, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada
| |
Collapse
|
4
|
Imamura H, Adachi T, Zhu W, Yamamoto T, Kanamura N, Onoda H, Nakamura-Takahashi A, Kasahara M, Nakada M, Sato H, Pezzotti G. Raman Spectroscopic Analysis of Molecular Structure and Mechanical Properties of Hypophosphatasia Primary Tooth. Molecules 2024; 29:6049. [PMID: 39770137 PMCID: PMC11678008 DOI: 10.3390/molecules29246049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/18/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Mild hypophosphatasia (HPP) can be difficult to distinguish from other bone disorders in the absence of typical symptoms such as the premature loss of primary teeth. Therefore, this study aimed to analyze the crystallinity of hydroxyapatite (HAp) and the three-dimensional structure of collagen in HPP teeth at the molecular level and to search for new biomarkers of HPP. Raman spectroscopy was used to investigate the molecular structure, composition, and mechanical properties of primary teeth from healthy individuals and patients with HPP. The results showed that the crystallinity of HAp decreased and the carbonate apatite content increased in the region near the dentin-enamel junction (DEJ) of HPP primary teeth. X-ray diffraction (XRD) analyses confirmed a decrease in HAp crystallinity near the DEJ, and micro-computed tomography (CT) scanning revealed a decrease in mineral density in this region. These results suggest incomplete calcification in HPP primary dentin and may contribute to the development of diagnostic and therapeutic agents.
Collapse
Affiliation(s)
- Hayata Imamura
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan; (H.I.); (W.Z.)
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (T.Y.); (N.K.); (G.P.)
| | - Tetsuya Adachi
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (T.Y.); (N.K.); (G.P.)
- Department of Dentistry, Kyoto Prefectural Rehabilitation Hospital for Mentally and Physically Disabled, Naka Ashihara, Joyo 610-0113, Japan
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
| | - Wenliang Zhu
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan; (H.I.); (W.Z.)
| | - Toshiro Yamamoto
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (T.Y.); (N.K.); (G.P.)
| | - Narisato Kanamura
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (T.Y.); (N.K.); (G.P.)
| | - Hiroaki Onoda
- Department of Biomolecular Chemistry, Faculty of Science and Technology, Kyoto Prefectural University, 1-5, Shimogamo-nakaragi, Sakyo-ku, Kyoto 606-8522, Japan;
| | - Aki Nakamura-Takahashi
- Department of Pharmacology, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan; (A.N.-T.); (M.K.)
| | - Masataka Kasahara
- Department of Pharmacology, Tokyo Dental College, 2-9-18 Kandamisaki-cho, Chiyoda-ku, Tokyo 101-0061, Japan; (A.N.-T.); (M.K.)
| | - Masaru Nakada
- Toray Research Center, Inc., 2-11 Sonoyama 3-chome, Otsu 520-8567, Japan;
| | - Hideo Sato
- Department of Pediatric Dentistry, Kagoshima University Hospital, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan;
| | - Giuseppe Pezzotti
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (T.Y.); (N.K.); (G.P.)
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
- Biomedical Engineering Center, Kansai Medical University, 1-9-11 Shin-machi, Hirakata, Osaka 573-1191, Japan
- Department of Orthopedic Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
- Department of Molecular Science and Nanosystems, Ca’ Foscari University of Venice, Via Torino 155, 30172 Venice, Italy
| |
Collapse
|
5
|
Soria-Tobar L, Román-Valero L, Sebastián-Serrano Á, Aivar P, Álvarez-Castelao B, Díaz-Hernández M. Blockade of brain alkaline phosphatase efficiently reduces amyloid-β plaque burden and associated cognitive impairment. Alzheimers Res Ther 2024; 16:233. [PMID: 39438925 PMCID: PMC11494749 DOI: 10.1186/s13195-024-01600-x] [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: 07/24/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most prevalent neurodegenerative disease. Three new drugs for AD based on monoclonal antibodies against the amyloid-β peptide (Aβ) have recently been approved because they favor the reduction of the burden of senile plaque in the AD patient's brain. Nonetheless, both drugs have very limited applicability and benefits and show several side effects. These limitations invite us to find alternative strategies for treating patients with AD. Here, we explored whether tissue-nonspecific alkaline phosphatase (TNAP), an ectoenzyme upregulated in the brain of AD patients and whose inhibition has beneficial effects on tau-induced pathology, is also efficient in reducing senile plaque burden. METHODS To evaluate whether TNAP may reduce cerebral senile plaque loading and Aβ-related toxicity, we use both pharmacological and genetic approaches. We analyze postmortem samples from human AD patients, APP/PS1 mice (a mouse model that mimics amyloid pathology observed in AD patients) treated or not with TNAP inhibitors, and the newly generated transgenic mouse line, TNAP-deficient APP/PS1 mice. RESULTS For the first time, we describe that genetic or pharmacological blockade of TNAP effectively reduces senile plaque burden by promoting its clearance, which leads to amelioration of cognitive impairment caused by Aβ-induced toxicity. These beneficial effects of TNAP inhibition occur concomitantly with higher microglial recruitment toward the senile plaque and increased microglial phagocytic capacity of Aβ by a mechanism involving metalloprotease-depending osteopontin processing. In addition, we also found that TNAP blockade favors LRP1-mediated transport of Aβ through the BBB. CONCLUSIONS Here, we have shown that TNAP inhibition effectively reduces brain senile plaque burden and associated behavioral defects. Furthermore, given that we had previously reported that TNAP blockade also ameliorates Tau-induced neurotoxicity and increases lifespan of P301S tauopathy mouse model, we can state that TNAP blockade may be a novel and efficient therapy for treating patients with AD.
Collapse
Affiliation(s)
- Lucia Soria-Tobar
- Department of Biochemistry and Molecular Biology, Veterinary School, Complutense University of Madrid, Avda. Puerta de Hierro S/N, Madrid, 28040, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| | - Laura Román-Valero
- Department of Biochemistry and Molecular Biology, Veterinary School, Complutense University of Madrid, Avda. Puerta de Hierro S/N, Madrid, 28040, Spain
| | - Álvaro Sebastián-Serrano
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, IdISSC, Madrid, Spain
- Department of Biochemistry and Molecular Biology, Medical School, Complutense University of Madrid, Plaza Ramón y Cajal, S/N, Madrid, 28040, Spain
| | - Paloma Aivar
- Department of Biochemistry and Molecular Biology, Veterinary School, Complutense University of Madrid, Avda. Puerta de Hierro S/N, Madrid, 28040, Spain
| | - Beatriz Álvarez-Castelao
- Department of Biochemistry and Molecular Biology, Veterinary School, Complutense University of Madrid, Avda. Puerta de Hierro S/N, Madrid, 28040, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, IdISSC, Madrid, Spain
| | - Miguel Díaz-Hernández
- Department of Biochemistry and Molecular Biology, Veterinary School, Complutense University of Madrid, Avda. Puerta de Hierro S/N, Madrid, 28040, Spain.
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, IdISSC, Madrid, Spain.
| |
Collapse
|
6
|
Malaval L, Follet H, Farlay D, Gineyts E, Rizzo S, Thomas C, Maalouf M, Normand M, Burt-Pichat B, Bouleftour W, Vanden-Boscche A, Laroche N, Vico L. OPN, BSP, and Bone Quality-Structural, Biochemical, and Biomechanical Assessment in OPN -/-, BSP -/-, and DKO Mice. Calcif Tissue Int 2024; 115:63-77. [PMID: 38733411 DOI: 10.1007/s00223-024-01217-0] [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: 11/20/2023] [Accepted: 04/12/2024] [Indexed: 05/13/2024]
Abstract
Osteopontin (OPN) and Bone Sialoprotein (BSP), abundantly expressed by osteoblasts and osteoclasts, appear to have important, partly overlapping functions in bone. In gene-knockout (KO, -/-) models of either protein and their double (D)KO in the same CD1/129sv genetic background, we analyzed the morphology, matrix characteristics, and biomechanical properties of femur bone in 2 and 4 month old, male and female mice. OPN-/- mice display inconsistent, perhaps localized hypermineralization, while the BSP-/- are hypomineralized throughout ages and sexes, and the low mineralization of young DKO mice recovers with age. The higher contribution of primary bone remnants in OPN-/- shafts suggests a slow turnover, while their lower percentage in BSP-/- indicates rapid remodeling, despite FTIR-based evidence in this genotype of a high maturity of the mineralized matrix. In 3-point bending assays, OPN-/- bones consistently display higher Maximal Load, Work to Max. Load and in young mice Ultimate Stress, an intrinsic characteristic of the matrix. Young male and old female BSP-/- also display high Work to Max. Load along with low Ultimate Stress. Principal Component Analysis confirms the major role of morphological traits in mechanical competence, and evidences a grouping of the WT phenotype with the OPN-/- and of BSP-/- with DKO, driven by both structural and matrix parameters, suggesting that the presence or absence of BSP has the most profound effects on skeletal properties. Single or double gene KO of OPN and BSP thus have multiple distinct effects on skeletal phenotypes, confirming their importance in bone biology and their interplay in its regulation.
Collapse
Affiliation(s)
- Luc Malaval
- LBTO, Pôle Santé Nord - Faculté de Médecine, Inserm, U1059 Sainbiose, Rm 118, Université Jean Monnet, Mines St Etienne, 10 Chemin de La Marandière, St Priest en Jarez, F42270, Saint-Etienne, France.
| | - Hélène Follet
- Inserm, Université de Lyon, U1033 Lyos, F69372, Lyon, France
| | - Delphine Farlay
- Inserm, Université de Lyon, U1033 Lyos, F69372, Lyon, France
| | - Evelyne Gineyts
- Inserm, Université de Lyon, U1033 Lyos, F69372, Lyon, France
| | - Sebastien Rizzo
- Inserm, Université de Lyon, U1033 Lyos, F69372, Lyon, France
| | - Charlene Thomas
- Inserm, Université de Lyon, U1033 Lyos, F69372, Lyon, France
| | - Mathieu Maalouf
- LBTO, Pôle Santé Nord - Faculté de Médecine, Inserm, U1059 Sainbiose, Rm 118, Université Jean Monnet, Mines St Etienne, 10 Chemin de La Marandière, St Priest en Jarez, F42270, Saint-Etienne, France
| | - Myriam Normand
- LBTO, Pôle Santé Nord - Faculté de Médecine, Inserm, U1059 Sainbiose, Rm 118, Université Jean Monnet, Mines St Etienne, 10 Chemin de La Marandière, St Priest en Jarez, F42270, Saint-Etienne, France
| | | | - Wafa Bouleftour
- Centre Hospitalier Universitaire de Saint-Etienne, Centre de Cancérologie Universitaire, F42270, Saint Etienne, France
| | - Arnaud Vanden-Boscche
- LBTO, Pôle Santé Nord - Faculté de Médecine, Inserm, U1059 Sainbiose, Rm 118, Université Jean Monnet, Mines St Etienne, 10 Chemin de La Marandière, St Priest en Jarez, F42270, Saint-Etienne, France
| | - Norbert Laroche
- LBTO, Pôle Santé Nord - Faculté de Médecine, Inserm, U1059 Sainbiose, Rm 118, Université Jean Monnet, Mines St Etienne, 10 Chemin de La Marandière, St Priest en Jarez, F42270, Saint-Etienne, France
| | - Laurence Vico
- LBTO, Pôle Santé Nord - Faculté de Médecine, Inserm, U1059 Sainbiose, Rm 118, Université Jean Monnet, Mines St Etienne, 10 Chemin de La Marandière, St Priest en Jarez, F42270, Saint-Etienne, France
| |
Collapse
|
7
|
Luisetto R, Scanu A. The translational value of calcium pyrophosphate deposition disease experimental mouse models. Front Med (Lausanne) 2024; 11:1417318. [PMID: 38846138 PMCID: PMC11153760 DOI: 10.3389/fmed.2024.1417318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 05/13/2024] [Indexed: 06/09/2024] Open
Abstract
The deposition of calcium pyrophosphate (CPP) crystals in joint tissues causes acute and chronic arthritis that commonly affect the adult and elderly population. Experimental calcium pyrophosphate deposition disease (CPPD) models are divided into genetically modified models and crystal-induced inflammation models. The former do not reproduce phenotypes overlapping with the human disease, while in the latter, the direct injection of crystals into the ankles, dorsal air pouch or peritoneum constitutes a useful and reliable methodology that resembles the CPP induced-inflammatory condition in humans. The translational importance of the induced model is also strengthened by the fact that the key molecular and cellular mediators involved in inflammation are shared between humans and laboratory rodents. Although, in vivo models are indispensable tools for studying the pathogenesis of the CPPD and testing new therapies, their development is still at an early stage and major efforts are needed to address this issue. Here, we analyze the strenghts and limitations of each currently available CPPD in vivo model, and critically discuss their translational value.
Collapse
Affiliation(s)
- Roberto Luisetto
- Experimental Surgery Research Center, Department of Surgery, Oncology and Gastroenterology-DISCOG, University of Padova, Padova, Italy
| | - Anna Scanu
- Department of Women's and Children's Health-SDB, University of Padova, Padova, Italy
- Departement of Neuroscience-DNS, University of Padova, Padova, Italy
| |
Collapse
|
8
|
Mohamed FF, de Oliveira FA, Kinoshita Y, Yalamanchili RR, Eltilib LA, Andras NL, Narisawa S, Tani T, Chu EY, Millán JL, Foster BL. Dentoalveolar Alterations in an Adenine-Induced Chronic Kidney Disease Mouse Model. J Bone Miner Res 2023; 38:1192-1207. [PMID: 37191192 PMCID: PMC10524958 DOI: 10.1002/jbmr.4829] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/02/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
Chronic kidney disease (CKD) is characterized by kidney damage and loss of renal function. CKD mineral and bone disorder (CKD-MBD) describes the dysregulation of mineral homeostasis, including hyperphosphatemia and elevated parathyroid hormone (PTH) secretion, skeletal abnormalities, and vascular calcification. CKD-MBD impacts the oral cavity, with effects including salivary gland dysfunction, enamel hypoplasia and damage, increased dentin formation, decreased pulp volume, pulp calcifications, and altered jaw bones, contributing to clinical manifestations of periodontal disease and tooth loss. Underlying mechanisms are not fully understood, and CKD mouse models commonly require invasive procedures with high rates of infection and mortality. We aimed to characterize the dentoalveolar effects of an adenine diet (AD)-induced CKD (AD-CKD) mouse model. Eight-week-old C57BL/6J mice were provided either a normal phosphorus diet control (CTR) or adenine and high-phosphorus diet CKD to induce kidney failure. Mice were euthanized at 15 weeks old, and mandibles were collected for micro-computed tomography and histology. CKD mice exhibited kidney failure, hyperphosphatemia, and hyperparathyroidism in association with porous cortical bone in femurs. CKD mice showed a 30% decrease in molar enamel volume compared to CTR mice. Enamel wear was associated with reduced ductal components, ectopic calcifications, and altered osteopontin (OPN) deposition in submandibular salivary glands of CKD mice. Molar cusps in CKD mice were flattened, exposing dentin. Molar dentin/cementum volume increased 7% in CKD mice and pulp volume decreased. Histology revealed excessive reactionary dentin and altered pulp-dentin extracellular matrix proteins, including increased OPN. Mandibular bone volume fraction decreased 12% and bone mineral density decreased 9% in CKD versus CTR mice. Alveolar bone in CKD mice exhibited increased tissue-nonspecific alkaline phosphatase localization, OPN deposition, and greater osteoclast numbers. AD-CKD recapitulated key aspects reported in CKD patients and revealed new insights into CKD-associated oral defects. This model has potential for studying mechanisms of dentoalveolar defects or therapeutic interventions. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Fatma F. Mohamed
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Flavia Amadeu de Oliveira
- Sanford Children’s Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Yuka Kinoshita
- Sanford Children’s Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Riti R. Yalamanchili
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Leena A. Eltilib
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Natalie L. Andras
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Sonoko Narisawa
- Sanford Children’s Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Takashi Tani
- Department of Endocrinology, Metabolism and Nephrology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Emily Y. Chu
- Department of General Dentistry, Operative Division, University of Maryland School of Dentistry, Baltimore, Maryland, USA
| | - José Luis Millán
- Sanford Children’s Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Brian L. Foster
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
9
|
Andras NL, Mohamed FF, Chu EY, Foster BL. Between a rock and a hard place: Regulation of mineralization in the periodontium. Genesis 2022; 60:e23474. [PMID: 35460154 PMCID: PMC9492628 DOI: 10.1002/dvg.23474] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/08/2022] [Accepted: 04/09/2022] [Indexed: 12/30/2022]
Abstract
The periodontium supports and attaches teeth via mineralized and nonmineralized tissues. It consists of two, unique mineralized tissues, cementum and alveolar bone. In between these tissues, lies an unmineralized, fibrous periodontal ligament (PDL), which distributes occlusal forces, nourishes and invests teeth, and harbors progenitor cells for dentoalveolar repair. Many unanswered questions remain regarding periodontal biology. This review will focus on recent research providing insights into one enduring mystery: the precise regulation of the hard-soft tissue borders in the periodontium which define the interfaces of the cementum-PDL-alveolar bone structure. We will focus on advances in understanding the molecular mechanisms that maintain the unmineralized PDL "between a rock and a hard place" by regulating the mineralization of cementum and alveolar bone.
Collapse
Affiliation(s)
- Natalie L. Andras
- Biosciences Division, College of DentistryThe Ohio State UniversityColumbusOhioUSA
| | - Fatma F. Mohamed
- Biosciences Division, College of DentistryThe Ohio State UniversityColumbusOhioUSA
| | - Emily Y. Chu
- Division of Operative Dentistry, Department of General Dentistry, School of DentistryUniversity of MarylandBaltimoreMarylandUSA
| | - Brian L. Foster
- Biosciences Division, College of DentistryThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
10
|
Nagasaki K, Gavrilova O, Hajishengallis G, Somerman MJ. Does the RGD region of certain proteins affect metabolic activity? FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.974862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A better understanding of the role of mineralized tissues and their associated factors in governing whole-body metabolism should be of value toward informing clinical strategies to treat mineralized tissue and metabolic disorders, such as diabetes and obesity. This perspective provides evidence suggesting a role for the arginine-glycine-aspartic acid (RGD) region, a sequence identified in several proteins secreted by bone cells, as well as other cells, in modulating systemic metabolic activity. We focus on (a) two of the SIBLING (small integrin-binding ligand, N-linked glycoprotein) family genes/proteins, bone sialoprotein (BSP) and osteopontin (OPN), (b) insulin-like growth factor-binding protein-1 & 2 (IGFBP-1, IGFBP-2) and (c) developmental endothelial locus 1 (DEL1) and milk fat globule–EGF factor-8 (MFG-E8). In addition, for our readers to appreciate the mounting evidence that a multitude of bone secreted factors affect the activity of other tissues, we provide a brief overview of other proteins, to include fibroblast growth factor 23 (FGF23), phosphatase orphan 1 (PHOSPHO1), osteocalcin (OCN/BGLAP), tissue non-specific alkaline phosphatase (TNAP) and acidic serine aspartic-rich MEPE-associated motif (ASARM), along with known/suggested functions of these factors in influencing energy metabolism.
Collapse
|
11
|
Figueredo CA, Abdelhay N, Gibson MP. The Roles of SIBLING Proteins in Dental, Periodontal and Craniofacial Development. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.898802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The majority of dental, periodontal, and craniofacial tissues are derived from the neural crest cells and ectoderm. Neural crest stem cells are pluripotent, capable of differentiating into a variety of cells. These cells can include osteoblasts, odontoblasts, cementoblasts, chondroblasts, and fibroblasts which are responsible for forming some of the tissues of the oral and craniofacial complex. The hard tissue forming cells deposit a matrix composed of collagen and non-collagenous proteins (NCPs) that later undergoes mineralization. The NCPs play a role in the mineralization of collagen. One such category of NCPs is the small integrin-binding ligand, N-linked glycoprotein (SIBLING) family of proteins. This family is composed of dentin sialophosphosprotein (DSPP), osteopontin (OPN), dentin matrix protein 1 (DMP1), bone sialoprotein (BSP), and matrix extracellular phosphoglycoprotein (MEPE). The SIBLING family is known to have regulatory effects in the mineralization process of collagen fibers and the maturation of hydroxyapatite crystals. It is well established that SIBLING proteins have critical roles in tooth development. Recent literature has described the expression and role of SIBLING proteins in other areas of the oral and craniofacial complex as well. The objective of the present literature review is to summarize and discuss the different roles the SIBLING proteins play in the development of dental, periodontal, and craniofacial tissues.
Collapse
|
12
|
Nam HK, Emmanouil E, Hatch NE. Deletion of the Pyrophosphate Generating Enzyme ENPP1 Rescues Craniofacial Abnormalities in the TNAP−/− Mouse Model of Hypophosphatasia and Reveals FGF23 as a Marker of Phenotype Severity. FRONTIERS IN DENTAL MEDICINE 2022; 3. [PMID: 35909501 PMCID: PMC9336114 DOI: 10.3389/fdmed.2022.846962] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Hypophosphatasia is a rare heritable metabolic disorder caused by deficient Tissue Non-specific Alkaline Phosphatase (TNAP) enzyme activity. A principal function of TNAP is to hydrolyze the tissue mineralization inhibitor pyrophosphate. ENPP1 (Ectonucleotide Pyrophosphatase/Phosphodiesterase 1) is a primary enzymatic generator of pyrophosphate and prior results showed that elimination of ENPP1 rescued bone hypomineralization of skull, vertebral and long bones to different extents in TNAP null mice. Current TNAP enzyme replacement therapy alleviates skeletal, motor and cognitive defects but does not eliminate craniosynostosis in pediatric hypophosphatasia patients. To further understand mechanisms underlying craniosynostosis development in hypophosphatasia, here we sought to determine if craniofacial abnormalities including craniosynostosis and skull shape defects would be alleviated in TNAP null mice by genetic ablation of ENPP1. Results show that homozygous deletion of ENPP1 significantly diminishes the incidence of craniosynostosis and that skull shape abnormalities are rescued by hemi- or homozygous deletion of ENPP1 in TNAP null mice. Skull and long bone hypomineralization were also alleviated in TNAP−/−/ENPP1−/− compared to TNAP−/−/ENPP1+/+ mice, though loss of ENPP1 in combination with TNAP had different effects than loss of only TNAP on long bone trabeculae. Investigation of a relatively large cohort of mice revealed that the skeletal phenotypes of TNAP null mice were markedly variable. Because FGF23 circulating levels are known to be increased in ENPP1 null mice and because FGF23 influences bone, we measured serum intact FGF23 levels in the TNAP null mice and found that a subset of TNAP−/−/ENPP1+/+ mice exhibited markedly high serum FGF23. Serum FGF23 levels also correlated to mouse body measurements, the incidence of craniosynostosis, skull shape abnormalities and skull bone density and volume fraction. Together, our results demonstrate that balanced expression of TNAP and ENPP1 enzymes are essential for microstructure and mineralization of both skull and long bones, and for preventing craniosynostosis. The results also show that FGF23 rises in the TNAP−/− model of murine lethal hypophosphatasia. Future studies are required to determine if the rise in FGF23 is a cause, consequence, or marker of disease phenotype severity.
Collapse
|
13
|
Mohamed FF, Chavez MB, de Oliveira FA, Narisawa S, Farquharson C, Millán JL, Foster BL. Perspective on Dentoalveolar Manifestations Resulting From PHOSPHO1 Loss-of-Function: A Form of Pseudohypophosphatasia? FRONTIERS IN DENTAL MEDICINE 2022; 3:826387. [PMID: 36185572 PMCID: PMC9521815 DOI: 10.3389/fdmed.2022.826387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Mineralization of the skeleton occurs by several physicochemical and biochemical processes and mechanisms that facilitate the deposition of hydroxyapatite (HA) in specific areas of the extracellular matrix (ECM). Two key phosphatases, phosphatase, orphan 1 (PHOSPHO1) and tissue-non-specific alkaline phosphatase (TNAP), play complementary roles in the mineralization process. The actions of PHOSPHO1 on phosphocholine and phosphoethanolamine in matrix vesicles (MVs) produce inorganic phosphate (Pi) for the initiation of HA mineral formation within MVs. TNAP hydrolyzes adenosine triphosphate (ATP) and the mineralization inhibitor, inorganic pyrophosphate (PPi), to generate Pi that is incorporated into MVs. Genetic mutations in the ALPL gene-encoding TNAP lead to hypophosphatasia (HPP), characterized by low circulating TNAP levels (ALP), rickets in children and/or osteomalacia in adults, and a spectrum of dentoalveolar defects, the most prevalent being lack of acellular cementum leading to premature tooth loss. Given that the skeletal manifestations of genetic ablation of the Phospho1 gene in mice resemble many of the manifestations of HPP, we propose that Phospho1 gene mutations may underlie some cases of "pseudo-HPP" where ALP may be normal to subnormal, but ALPL mutation(s) have not been identified. The goal of this perspective article is to compare and contrast the loss-of-function effects of TNAP and PHOSPHO1 on the dentoalveolar complex to predict the likely dental phenotype in humans that may result from PHOSPHO1 mutations. Potential cases of pseudo-HPP associated with PHOSPHO1 mutations may resist diagnosis, and the dental manifestations could be a key criterion for consideration.
Collapse
Affiliation(s)
- Fatma F. Mohamed
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Michael B. Chavez
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Flavia Amadeu de Oliveira
- Sanford Children’s Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Sonoko Narisawa
- Sanford Children’s Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Colin Farquharson
- The Royal (Dick) School of Veterinary Studies (RDSVS), The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - José Luis Millán
- Sanford Children’s Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Brian L. Foster
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States,,Correspondence: Brian L. Foster,
| |
Collapse
|
14
|
Saxena A, Sharma V, Muthuirulan P, Neufeld SJ, Tran MP, Gutierrez HL, Chen KD, Erberich JM, Birmingham A, Capellini TD, Cobb J, Hiller M, Cooper KL. Interspecies transcriptomics identify genes that underlie disproportionate foot growth in jerboas. Curr Biol 2022; 32:289-303.e6. [PMID: 34793695 PMCID: PMC8792248 DOI: 10.1016/j.cub.2021.10.063] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 07/16/2021] [Accepted: 10/28/2021] [Indexed: 01/26/2023]
Abstract
Despite the great diversity of vertebrate limb proportion and our deep understanding of the genetic mechanisms that drive skeletal elongation, little is known about how individual bones reach different lengths in any species. Here, we directly compare the transcriptomes of homologous growth cartilages of the mouse (Mus musculus) and bipedal jerboa (Jaculus jaculus), the latter of which has "mouse-like" arms but extremely long metatarsals of the feet. Intersecting gene-expression differences in metatarsals and forearms of the two species revealed that about 10% of orthologous genes are associated with the disproportionately rapid elongation of neonatal jerboa feet. These include genes and enriched pathways not previously associated with endochondral elongation as well as those that might diversify skeletal proportion in addition to their known requirements for bone growth throughout the skeleton. We also identified transcription regulators that might act as "nodes" for sweeping differences in genome expression between species. Among these, Shox2, which is necessary for proximal limb elongation, has gained expression in jerboa metatarsals where it has not been detected in other vertebrates. We show that Shox2 is sufficient to increase mouse distal limb length, and a nearby putative cis-regulatory region is preferentially accessible in jerboa metatarsals. In addition to mechanisms that might directly promote growth, we found evidence that jerboa foot elongation may occur in part by de-repressing latent growth potential. The genes and pathways that we identified here provide a framework to understand the modular genetic control of skeletal growth and the remarkable malleability of vertebrate limb proportion.
Collapse
Affiliation(s)
- Aditya Saxena
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Virag Sharma
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, Dresden 01307, Germany; Max Planck Institute for the Physics of Complex Systems, Nothnitzerstraße 38, Dresden 01187, Germany
| | - Pushpanathan Muthuirulan
- Department of Human Evolutionary Biology, Harvard University, 11 Divinity Avenue, Cambridge, MA 02138, USA
| | - Stanley J Neufeld
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Mai P Tran
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Haydee L Gutierrez
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kevin D Chen
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Joel M Erberich
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Amanda Birmingham
- Center for Computational Biology and Bioinformatics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Terence D Capellini
- Department of Human Evolutionary Biology, Harvard University, 11 Divinity Avenue, Cambridge, MA 02138, USA
| | - John Cobb
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| | - Michael Hiller
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, Dresden 01307, Germany; Max Planck Institute for the Physics of Complex Systems, Nothnitzerstraße 38, Dresden 01187, Germany
| | - Kimberly L Cooper
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
15
|
Goettsch C, Strzelecka-Kiliszek A, Bessueille L, Quillard T, Mechtouff L, Pikula S, Canet-Soulas E, Luis MJ, Fonta C, Magne D. TNAP as a therapeutic target for cardiovascular calcification: a discussion of its pleiotropic functions in the body. Cardiovasc Res 2022; 118:84-96. [PMID: 33070177 PMCID: PMC8752354 DOI: 10.1093/cvr/cvaa299] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/11/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular calcification (CVC) is associated with increased morbidity and mortality. It develops in several diseases and locations, such as in the tunica intima in atherosclerosis plaques, in the tunica media in type 2 diabetes and chronic kidney disease, and in aortic valves. In spite of the wide occurrence of CVC and its detrimental effects on cardiovascular diseases (CVD), no treatment is yet available. Most of CVC involve mechanisms similar to those occurring during endochondral and/or intramembranous ossification. Logically, since tissue-nonspecific alkaline phosphatase (TNAP) is the key-enzyme responsible for skeletal/dental mineralization, it is a promising target to limit CVC. Tools have recently been developed to inhibit its activity and preclinical studies conducted in animal models of vascular calcification already provided promising results. Nevertheless, as its name indicates, TNAP is ubiquitous and recent data indicate that it dephosphorylates different substrates in vivo to participate in other important physiological functions besides mineralization. For instance, TNAP is involved in the metabolism of pyridoxal phosphate and the production of neurotransmitters. TNAP has also been described as an anti-inflammatory enzyme able to dephosphorylate adenosine nucleotides and lipopolysaccharide. A better understanding of the full spectrum of TNAP's functions is needed to better characterize the effects of TNAP inhibition in diseases associated with CVC. In this review, after a brief description of the different types of CVC, we describe the newly uncovered additional functions of TNAP and discuss the expected consequences of its systemic inhibition in vivo.
Collapse
Affiliation(s)
- Claudia Goettsch
- Department of Internal Medicine I, Cardiology, Medical Faculty, RWTH Aachen
University, Aachen, Germany
| | - Agnieszka Strzelecka-Kiliszek
- Laboratory of Biochemistry of Lipids, Nencki Institute of Experimental
Biology, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Laurence Bessueille
- Institute of Molecular and Supramolecular Chemistry and Biochemistry
(ICBMS), UMR CNRS 5246, Université Claude Bernard Lyon 1, Bâtiment
Raulin, 43 Bd du 11 novembre 1918, Lyon 69622 Villeurbanne Cedex, France
| | - Thibaut Quillard
- PHY-OS Laboratory, UMR 1238 INSERM, Université de Nantes, CHU
de Nantes, France
| | - Laura Mechtouff
- Stroke Department, Hospices Civils de Lyon, France
- CREATIS Laboratory, CNRS UMR 5220, Inserm U1044, Université Claude Bernard
Lyon 1, Lyon, France
| | - Slawomir Pikula
- Laboratory of Biochemistry of Lipids, Nencki Institute of Experimental
Biology, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Emmanuelle Canet-Soulas
- CarMeN Laboratory, Univ Lyon, INSERM, INRA, INSA Lyon, Université Claude
Bernard Lyon 1, Lyon, France
| | - Millan Jose Luis
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery
Institute, La Jolla, CA 92037, USA
| | - Caroline Fonta
- Brain and Cognition Research Center CerCo, CNRS UMR5549, Université de
Toulouse, France
| | - David Magne
- Institute of Molecular and Supramolecular Chemistry and Biochemistry
(ICBMS), UMR CNRS 5246, Université Claude Bernard Lyon 1, Bâtiment
Raulin, 43 Bd du 11 novembre 1918, Lyon 69622 Villeurbanne Cedex, France
| |
Collapse
|
16
|
Sekaran S, Vimalraj S, Thangavelu L. The Physiological and Pathological Role of Tissue Nonspecific Alkaline Phosphatase beyond Mineralization. Biomolecules 2021; 11:1564. [PMID: 34827562 PMCID: PMC8615537 DOI: 10.3390/biom11111564] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/12/2021] [Accepted: 10/12/2021] [Indexed: 12/17/2022] Open
Abstract
Tissue-nonspecific alkaline phosphatase (TNAP) is a key enzyme responsible for skeletal tissue mineralization. It is involved in the dephosphorylation of various physiological substrates, and has vital physiological functions, including extra-skeletal functions, such as neuronal development, detoxification of lipopolysaccharide (LPS), an anti-inflammatory role, bile pH regulation, and the maintenance of the blood brain barrier (BBB). TNAP is also implicated in ectopic pathological calcification of soft tissues, especially the vasculature. Although it is the crucial enzyme in mineralization of skeletal and dental tissues, it is a logical clinical target to attenuate vascular calcification. Various tools and studies have been developed to inhibit its activity to arrest soft tissue mineralization. However, we should not neglect its other physiological functions prior to therapies targeting TNAP. Therefore, a better understanding into the mechanisms mediated by TNAP is needed for minimizing off targeted effects and aid in the betterment of various pathological scenarios. In this review, we have discussed the mechanism of mineralization and functions of TNAP beyond its primary role of hard tissue mineralization.
Collapse
Affiliation(s)
- Saravanan Sekaran
- Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospitals, Saveetha University, Chennai 600 077, Tamil Nadu, India;
| | - Selvaraj Vimalraj
- Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospitals, Saveetha University, Chennai 600 077, Tamil Nadu, India;
- Centre for Biotechnology, Anna University, Chennai 600 025, Tamil Nadu, India
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospitals, Saveetha University, Chennai 600 077, Tamil Nadu, India;
| |
Collapse
|
17
|
Nagasaki A, Nagasaki K, Chu EY, Kear BD, Tadesse WD, Ferebee SE, Li L, Foster BL, Somerman MJ. Ablation of Pyrophosphate Regulators Promotes Periodontal Regeneration. J Dent Res 2020; 100:639-647. [PMID: 33356859 DOI: 10.1177/0022034520981854] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Biomineralization is regulated by inorganic pyrophosphate (PPi), a potent physiological inhibitor of hydroxyapatite crystal growth. Progressive ankylosis protein (ANK) and ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) act to increase local extracellular levels of PPi, inhibiting mineralization. The periodontal complex includes 2 mineralized tissues, cementum and alveolar bone (AB), both essential for tooth attachment. Previous studies demonstrated that loss of function of ANK or ENPP1 (reducing PPi) resulted in increased cementum formation, suggesting PPi metabolism may be a target for periodontal regenerative therapies. To compare the effects of genetic ablation of Ank, Enpp1, and both factors concurrently on cementum and AB regeneration, mandibular fenestration defects were created in Ank knockout (Ank KO), Enpp1 mutant (Enpp1asj/asj), and double KO (dKO) mice. Genetic ablation of Ank, Enpp1, or both factors increased cementum regeneration compared to controls at postoperative days (PODs) 15 and 30 (Ank KO: 8-fold, 3-fold; Enpp1asj/asj: 7-fold, 3-fold; dKO: 11-fold, 4-fold, respectively) associated with increased fluorochrome labeling and expression of mineralized tissue markers, dentin matrix protein 1 (Dmp1/DMP1), osteopontin (Spp1/OPN), and bone sialoprotein (Ibsp/BSP). Furthermore, dKO mice featured increased cementum thickness compared to single KOs at POD15 and Ank KO at POD30. No differences were noted in AB volume between genotypes, but osteoblast/osteocyte markers were increased in all KOs, partially mineralized osteoid volume was increased in dKO versus controls at POD15 (3-fold), and mineral density was decreased in Enpp1asj/asj and dKOs at POD30 (6% and 9%, respectively). Increased numbers of osteoclasts were present in regenerated AB of all KOs versus controls. These preclinical studies suggest PPi modulation as a potential and novel approach for cementum regeneration, particularly targeting ENPP1 and/or ANK. Differences in cementum and AB regeneration in response to reduced PPi conditions highlight the need to consider tissue-specific responses in strategies targeting regeneration of the entire periodontal complex.
Collapse
Affiliation(s)
- A Nagasaki
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - K Nagasaki
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - E Y Chu
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - B D Kear
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - W D Tadesse
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - S E Ferebee
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - L Li
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - B L Foster
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - M J Somerman
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
18
|
Bailey S, Sroga GE, Hoac B, Katsamenis OL, Wang Z, Bouropoulos N, McKee MD, Sørensen ES, Thurner PJ, Vashishth D. The role of extracellular matrix phosphorylation on energy dissipation in bone. eLife 2020; 9:58184. [PMID: 33295868 PMCID: PMC7746230 DOI: 10.7554/elife.58184] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 12/07/2020] [Indexed: 01/22/2023] Open
Abstract
Protein phosphorylation, critical for cellular regulatory mechanisms, is implicated in various diseases. However, it remains unknown whether heterogeneity in phosphorylation of key structural proteins alters tissue integrity and organ function. Here, osteopontin phosphorylation level declined in hypo- and hyper- phosphatemia mouse models exhibiting skeletal deformities. Phosphorylation increased cohesion between osteopontin polymers, and adhesion of osteopontin to hydroxyapatite, enhancing energy dissipation. Fracture toughness, a measure of bone’s mechanical competence, increased with ex-vivo phosphorylation of wildtype mouse bones and declined with ex-vivo dephosphorylation. In osteopontin-deficient mice, global matrix phosphorylation level was not associated with toughness. Our findings suggest that phosphorylated osteopontin promotes fracture toughness in a dose-dependent manner through increased interfacial bond formation. In the absence of osteopontin, phosphorylation increases electrostatic repulsion, and likely protein alignment and interfilament distance leading to decreased fracture resistance. These mechanisms may be of importance in other connective tissues, and the key to unraveling cell–matrix interactions in diseases.
Collapse
Affiliation(s)
- Stacyann Bailey
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, United States
| | - Grazyna E Sroga
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, United States
| | - Betty Hoac
- Faculty of Dentistry, McGill University, Montreal, Canada
| | - Orestis L Katsamenis
- Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, United Kingdom
| | - Zehai Wang
- Department of Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, United States
| | | | - Marc D McKee
- Faculty of Dentistry, McGill University, Montreal, Canada.,Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, Canada
| | - Esben S Sørensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Philipp J Thurner
- Institute of Lightweight Design and Structural Biomechanics, Vienna University of Technology, Vienna, Austria
| | - Deepak Vashishth
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, United States
| |
Collapse
|
19
|
Tissue-Nonspecific Alkaline Phosphatase-A Gatekeeper of Physiological Conditions in Health and a Modulator of Biological Environments in Disease. Biomolecules 2020; 10:biom10121648. [PMID: 33302551 PMCID: PMC7763311 DOI: 10.3390/biom10121648] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 12/15/2022] Open
Abstract
Tissue-nonspecific alkaline phosphatase (TNAP) is a ubiquitously expressed enzyme that is best known for its role during mineralization processes in bones and skeleton. The enzyme metabolizes phosphate compounds like inorganic pyrophosphate and pyridoxal-5′-phosphate to provide, among others, inorganic phosphate for the mineralization and transportable vitamin B6 molecules. Patients with inherited loss of function mutations in the ALPL gene and consequently altered TNAP activity are suffering from the rare metabolic disease hypophosphatasia (HPP). This systemic disease is mainly characterized by impaired bone and dental mineralization but may also be accompanied by neurological symptoms, like anxiety disorders, seizures, and depression. HPP characteristically affects all ages and shows a wide range of clinical symptoms and disease severity, which results in the classification into different clinical subtypes. This review describes the molecular function of TNAP during the mineralization of bones and teeth, further discusses the current knowledge on the enzyme’s role in the nervous system and in sensory perception. An additional focus is set on the molecular role of TNAP in health and on functional observations reported in common laboratory vertebrate disease models, like rodents and zebrafish.
Collapse
|
20
|
Hoac B, Østergaard M, Wittig NK, Boukpessi T, Buss DJ, Chaussain C, Birkedal H, Murshed M, McKee MD. Genetic Ablation of Osteopontin in Osteomalacic Hyp Mice Partially Rescues the Deficient Mineralization Without Correcting Hypophosphatemia. J Bone Miner Res 2020; 35:2032-2048. [PMID: 32501585 DOI: 10.1002/jbmr.4101] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/24/2020] [Accepted: 06/01/2020] [Indexed: 11/09/2022]
Abstract
PHEX is predominantly expressed by bone and tooth-forming cells, and its inactivating mutations in X-linked hypophosphatemia (XLH) lead to renal phosphate wasting and severe hypomineralization of bones and teeth. Also present in XLH are hallmark hypomineralized periosteocytic lesions (POLs, halos) that persist despite stable correction of serum phosphate (Pi ) that improves bulk bone mineralization. In XLH, mineralization-inhibiting osteopontin (OPN, a substrate for PHEX) accumulates in the extracellular matrix of bone. To investigate how OPN functions in Hyp mice (a model for XLH), double-null (Hyp;Opn-/- ) mice were generated. Undecalcified histomorphometry performed on lumbar vertebrae revealed that Hyp;Opn-/- mice had significantly reduced osteoid area/bone area (OV/BV) and osteoid thickness of trabecular bone as compared to Hyp mice, despite being as hypophosphatemic as Hyp littermate controls. However, tibias examined by synchrotron radiation micro-CT showed that mineral lacunar volumes remained abnormally enlarged in these double-null mice. When Hyp;Opn-/- mice were fed a high-Pi diet, serum Pi concentration increased, and OV/BV and osteoid thickness normalized, yet mineral lacunar area remained abnormally enlarged. Enpp1 and Ankh gene expression were increased in double-null mice fed a high-Pi diet, potentially indicating a role for elevated inhibitory pyrophosphate (PPi ) in the absence of OPN. To further investigate the persistence of POLs in Hyp mice despite stable correction of serum Pi , immunohistochemistry for OPN on Hyp mice fed a high-Pi diet showed elevated OPN in the osteocyte pericellular lacunar matrix as compared to Hyp mice fed a control diet. This suggests that POLs persisting in Hyp mice despite correction of serum Pi may be attributable to the well-known upregulation of mineralization-inhibiting OPN by Pi , and its accumulation in the osteocyte pericellular matrix. This study shows that OPN contributes to osteomalacia in Hyp mice, and that genetic ablation of OPN in Hyp mice improves the mineralization phenotype independent of systemic Pi -regulating factors. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Betty Hoac
- Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - Maja Østergaard
- Department of Chemistry and the Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark
| | - Nina K Wittig
- Department of Chemistry and the Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark
| | - Tchilalo Boukpessi
- Faculty of Dentistry, McGill University, Montreal, QC, Canada.,EA 2496, Laboratory Orofacial Pathologies, Imaging and Biotherapies, School of Dentistry Université de Paris, Paris, France.,Assistance Publique-Hôpitaux de Paris (AP-HP) Department of Odontology, Charles Foix and Bretonneau Hospitals and Reference Center for Rare Diseases of Calcium and Phosphorus Metabolism, Paris, France
| | - Daniel J Buss
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Catherine Chaussain
- EA 2496, Laboratory Orofacial Pathologies, Imaging and Biotherapies, School of Dentistry Université de Paris, Paris, France.,Assistance Publique-Hôpitaux de Paris (AP-HP) Department of Odontology, Charles Foix and Bretonneau Hospitals and Reference Center for Rare Diseases of Calcium and Phosphorus Metabolism, Paris, France
| | - Henrik Birkedal
- Department of Chemistry and the Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark
| | - Monzur Murshed
- Faculty of Dentistry, McGill University, Montreal, QC, Canada.,Department of Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada.,Shriners Hospital for Children, Montreal, QC, Canada
| | - Marc D McKee
- Faculty of Dentistry, McGill University, Montreal, QC, Canada.,Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
21
|
Reznikov N, Hoac B, Buss DJ, Addison WN, Barros NMT, McKee MD. Biological stenciling of mineralization in the skeleton: Local enzymatic removal of inhibitors in the extracellular matrix. Bone 2020; 138:115447. [PMID: 32454257 DOI: 10.1016/j.bone.2020.115447] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/14/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022]
Abstract
Biomineralization is remarkably diverse and provides myriad functions across many organismal systems. Biomineralization processes typically produce hardened, hierarchically organized structures usually having nanostructured mineral assemblies that are formed through inorganic-organic (usually protein) interactions. Calcium‑carbonate biomineral predominates in structures of small invertebrate organisms abundant in marine environments, particularly in shells (remarkably it is also found in the inner ear otoconia of vertebrates), whereas calcium-phosphate biomineral predominates in the skeletons and dentitions of both marine and terrestrial vertebrates, including humans. Reconciliation of the interplay between organic moieties and inorganic crystals in bones and teeth is a cornerstone of biomineralization research. Key molecular determinants of skeletal and dental mineralization have been identified in health and disease, and in pathologic ectopic calcification, ranging from small molecules such as pyrophosphate, to small membrane-bounded matrix vesicles shed from cells, and to noncollagenous extracellular matrix proteins such as osteopontin and their derived bioactive peptides. Beyond partly knowing the regulatory role of the direct actions of inhibitors on vertebrate mineralization, more recently the importance of their enzymatic removal from the extracellular matrix has become increasingly understood. Great progress has been made in deciphering the relationship between mineralization inhibitors and the enzymes that degrade them, and how adverse changes in this physiologic pathway (such as gene mutations causing disease) result in mineralization defects. Two examples of this are rare skeletal diseases having osteomalacia/odontomalacia (soft bones and teeth) - namely hypophosphatasia (HPP) and X-linked hypophosphatemia (XLH) - where inactivating mutations occur in the gene for the enzymes tissue-nonspecific alkaline phosphatase (TNAP, TNSALP, ALPL) and phosphate-regulating endopeptidase homolog X-linked (PHEX), respectively. Here, we review and provide a concept for how existing and new information now comes together to describe the dual nature of regulation of mineralization - through systemic mineral ion homeostasis involving circulating factors, coupled with molecular determinants operating at the local level in the extracellular matrix. For the local mineralization events in the extracellular matrix, we present a focused concept in skeletal mineralization biology called the Stenciling Principle - a principle (building upon seminal work by Neuman and Fleisch) describing how the action of enzymes to remove tissue-resident inhibitors defines with precision the location and progression of mineralization.
Collapse
Affiliation(s)
- N Reznikov
- Object Research Systems Inc., 760 St. Paul West, Montreal, Quebec H3C 1M4, Canada.
| | - B Hoac
- Faculty of Dentistry, McGill University, 3640 University St., Montreal, Quebec H3A 0C7, Canada
| | - D J Buss
- Department of Anatomy and Cell Biology, McGill University, 3640 University St., Montreal, Quebec H3A 0C7, Canada
| | - W N Addison
- Department of Molecular Signaling and Biochemistry, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka, Japan
| | - N M T Barros
- Departamento de Biofísica, São Paulo, Departamento de Ciências Biológicas, Universidade Federal de São Paulo, Diadema, Brazil
| | - M D McKee
- Faculty of Dentistry, McGill University, 3640 University St., Montreal, Quebec H3A 0C7, Canada; Department of Anatomy and Cell Biology, McGill University, 3640 University St., Montreal, Quebec H3A 0C7, Canada.
| |
Collapse
|
22
|
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
|
23
|
Naqvi SM, Panadero Pérez JA, Kumar V, Verbruggen ASK, McNamara LM. A Novel 3D Osteoblast and Osteocyte Model Revealing Changes in Mineralization and Pro-osteoclastogenic Paracrine Signaling During Estrogen Deficiency. Front Bioeng Biotechnol 2020; 8:601. [PMID: 32656194 PMCID: PMC7326002 DOI: 10.3389/fbioe.2020.00601] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/18/2020] [Indexed: 11/13/2022] Open
Abstract
Recent in vitro studies have revealed that the mechanobiological responses of osteoblasts and osteocytes are fundamentally impaired during estrogen deficiency. However, these two-dimensional (2D) cell culture studies do not account for in vivo biophysical cues. Thus, the objectives of this study are to (1) develop a three-dimensional (3D) osteoblast and osteocyte model integrated into a bioreactor and (2) apply this model to investigate whether estrogen deficiency leads to changes in osteoblast to osteocyte transition, mechanosensation, mineralization, and paracrine signaling associated with bone resorption by osteoclasts. MC3T3-E1s were expanded in media supplemented with estrogen (17β-estradiol). These cells were encapsulated in gelatin-mtgase before culture in (1) continued estrogen (E) or (2) no further estrogen supplementation. Constructs were placed in gas permeable and water impermeable cell culture bags and maintained at 5% CO2 and 37°C. These bags were either mechanically stimulated in a custom hydrostatic pressure (HP) bioreactor or maintained under static conditions (control). We report that osteocyte differentiation, characterized by the presence of dendrites and staining for osteocyte marker dentin matrix acidic phosphoprotein 1 (DMP1), was significantly greater under estrogen withdrawal (EW) compared to under continuous estrogen treatment (day 21). Mineralization [bone sialoprotein (BSP), osteopontin (OPN), alkaline phosphatase (ALP), calcium] and gene expression associated with paracrine signaling for osteoclastogenesis [receptor activator of nuclear factor kappa-β ligand (RANKL)/osteoprotegerin OPG ratio] were significantly increased in estrogen deficient and mechanically stimulated cells. Interestingly, BSP and DMP-1 were also increased at day 1 and day 21, respectively, which play a role in regulation of biomineralization. Furthermore, the increase in pro-osteoclastogenic signaling may be explained by altered mechanoresponsiveness of osteoblasts or osteocytes during EW. These findings highlight the impact of estrogen deficiency on bone cell function and provide a novel in vitro model to investigate the mechanisms underpinning changes in bone cells after estrogen deficiency.
Collapse
Affiliation(s)
| | | | | | | | - Laoise M. McNamara
- Mechanobiology and Medical Device Research Group, Department of Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
24
|
Synthesis and computational studies of highly selective inhibitors of human recombinant tissue non-specific alkaline phosphatase (h-TNAP): A therapeutic target against vascular calcification. Bioorg Chem 2020; 101:103999. [PMID: 32563966 DOI: 10.1016/j.bioorg.2020.103999] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022]
Abstract
In this study, we have discovered small druglike molecules as selective inhibitors of human tissue-nonspecific alkaline phosphatase (h-TNAP), an enzyme critical for the regulation of extracellular matrix calcification. The upregulation of h-TNAP is associated with various pathologies particularly the vascular calcification (VC). Selective inhibition of h-TNAP over h-NPP1 may serve as a useful therapeutic strategy against vascular calcification. A series of novel triazolyl pyrazole derivatives (10a-y) in which thiol bearing triazole moiety as the zinc binding functional group was introduced to a pyrazole based pharmacophore was synthesized and evaluated as potent and selective inhibitors of h-TNAP over h-NPP1. The biological screening against h-TNAP, h-IAP, h-NPP1 and h-NPP3 showed that many of the synthesized compounds are selective inhibitors of TNAP. Particularly, the compounds 10a-h, 10j, 10m-q, 10u, 10w and 10x displayed high potency and complete selectivity towards h-TNAP over h-NPP1. Compound 10q emerged as a highly potent inhibitor (IC50 = 0.16 µM or 160 nM) against h-TNAP with 127-fold increased inhibition compared to levamisole. On the other hand, compound 10e was found to be most selective inhibitor against the tested APs and NPPs (IC50 = 1.59 ± 0.36 µM). Binding sites architecture analysis, molecular-docking and molecular dynamics simulations (MDS), revealed the basis for h-TNAP and h-IAP ligand selectivity as well as selectivity towards h-TNAP over h-NPP1. These newly discovered inhibitors are believed to represent valuable lead structures to further streamline the generation of candidate compounds to target VC.
Collapse
|
25
|
Melms H, Herrmann M, Förstner K, Bharti R, Schneider D, Mentrup B, Rudert M, Schlagenhauf U, Jakob F, Graser S. Novel molecular cues for dental defects in hypophosphatasia. Exp Cell Res 2020; 392:112026. [PMID: 32333908 DOI: 10.1016/j.yexcr.2020.112026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 01/09/2023]
Abstract
Mineralization disorders with a broad range of etiological factors represent a huge challenge in dental diagnosis and therapy. Hypophosphatasia (HPP) belongs to the rare diseases affecting predominantly mineralized tissues, bones and teeth, and occurs due to mutations in the ALPL gene, which encodes tissue-nonspecific alkaline phosphatase (TNAP). Here we analyzed stem cells from bone marrow (BMSCs), dental pulp (DPSCs) and periodontal ligament (PDLSCs) in the absence and presence of efficient TNAP inhibitors. The differentiation capacity, expression of surface markers, and gene expression patterns of donor-matched dental cells were compared during this in vitro study. Differentiation assays showed efficient osteogenic but low adipogenic differentiation (aD) capacity of PDLSCs and DPSCs. TNAP inhibitor treatment completely abolished the mineralization process during osteogenic differentiation (oD). RNA-seq analysis in PDLSCs, comparing oD with and without TNAP inhibitor levamisole, showed clustered regulation of candidate molecular mechanisms that putatively impaired osteogenesis and mineralization, disequilibrated ECM production and turnover, and propagated inflammation. Combined alteration of cementum formation, mineralization, and elastic attachment of teeth to cementum via elastic fibers may explain dental key problems in HPP. Using this in vitro model of TNAP deficiency in DPSCs and PDLSCs, we provide novel putative target areas for research on molecular cues for specific dental problems in HPP.
Collapse
Affiliation(s)
- Hannah Melms
- Bernhard-Heine-Center for Locomotion Research, University of Würzburg, Würzburg, Germany; Department of Conservative Dentistry, School of Dental Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Marietta Herrmann
- Bernhard-Heine-Center for Locomotion Research, University of Würzburg, Würzburg, Germany; IZKF Research Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Würzburg, Würzburg, Germany
| | - Konrad Förstner
- Core Unit SysMed, University of Würzburg, Würzburg, Germany; Institute for Information Sciences, University of Applied Sciences, Cologne, Germany; ZB MED - Information Centre for Life Science, Cologne, Germany
| | - Richa Bharti
- Core Unit SysMed, University of Würzburg, Würzburg, Germany; Department for Bioinformatics, Weihenstephan-Triesdorf University of Applied Sciences, TUM Campus Straubing for Biotechnology and Sustainability, Straubing, Germany
| | - Doris Schneider
- Bernhard-Heine-Center for Locomotion Research, University of Würzburg, Würzburg, Germany
| | - Birgit Mentrup
- Bernhard-Heine-Center for Locomotion Research, University of Würzburg, Würzburg, Germany; Department for Regenerative Musculoskeletal Medicine, University of Münster, Germany
| | - Maximilian Rudert
- Department of Orthopaedics, Orthopedic Department König-Ludwig-Haus, University of Würzburg, Würzburg, Germany
| | - Ulrich Schlagenhauf
- Department of Periodontology, University Clinics Würzburg, Würzburg, Germany
| | - Franz Jakob
- Bernhard-Heine-Center for Locomotion Research, University of Würzburg, Würzburg, Germany
| | - Stephanie Graser
- Bernhard-Heine-Center for Locomotion Research, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
26
|
Abstract
Hypophosphatasia (HPP) is a rare inherited systemic metabolic disease caused by mutations in the tissue-nonspecific alkaline phosphatase (TNSALP) gene. TNSALP is expressed in the liver, kidney and bone, and its substrates include TNSALP inorganic pyrophosphate, pyridoxal-5'-phosphate (PLP)/vitamin B6 and phosphoethanolamine (PEA). Autosomal recessive and dominant forms of the disease result in a range of clinical entities. Major hallmarks are low alkaline phosphatase (ALP) and elevated PLP and PEA levels. Very severe infantile forms of HPP cause premature death as a result of respiratory insufficiency and also present with hypo-mineralisation leading to deformed limbs with, in some cases, the near-absence of bones and skull altogether. Respiratory failure, rib fractures and seizures due to vitamin B6 deficiency are indicative of a poor prognosis. Craniosynostosis is frequent. HPP leads to an unusual presentation of rickets with high levels of calcium and phosphorus, resulting in hypercalciuria, nephrocalcinosis and low ALP levels. Hypercalcaemic crisis, failure to thrive and growth retardation are concerns in infants. Fractures are common in both infantile and adult forms of the disease, concomitantly occurring with unexplained chronic pain and fatigue. Dental clinical presentations, which include the premature loss of teeth, are also commonly found in HPP and specifically manifest as odontohypophosphatasia. A novel enzyme therapy for human HPP, asfotase alfa, which is specifically targeted to mineralised tissues, has been developed in the past decades. While this treatment seems very promising, especially for infantile HPP, many questions regarding its long-term effects, the management of treatment, and any potential secondary adverse effects remain unresolved.
Collapse
|
27
|
Bowden SA, Foster BL. Alkaline Phosphatase Replacement Therapy for Hypophosphatasia in Development and Practice. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1148:279-322. [PMID: 31482504 DOI: 10.1007/978-981-13-7709-9_13] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Hypophosphatasia (HPP) is an inherited disorder that affects bone and tooth mineralization characterized by low serum alkaline phosphatase. HPP is caused by loss-of-function mutations in the ALPL gene encoding the protein, tissue-nonspecific alkaline phosphatase (TNSALP). TNSALP is expressed by mineralizing cells of the skeleton and dentition and is associated with the mineralization process. Generalized reduction of activity of the TNSALP leads to accumulation of its substrates, including inorganic pyrophosphate (PPi) that inhibits physiological mineralization. This leads to defective skeletal mineralization, with manifestations including rickets, osteomalacia, fractures, and bone pain, all of which can result in multi-systemic complications with significant morbidity, as well as mortality in severe cases. Dental manifestations are nearly universal among affected individuals and feature most prominently premature loss of deciduous teeth. Management of HPP has been limited to supportive care until the introduction of a TNSALP enzyme replacement therapy (ERT), asfotase alfa (AA). AA ERT has proven to be transformative, improving survival in severely affected infants and increasing overall quality of life in children and adults with HPP. This chapter provides an overview of TNSALP expression and functions, summarizes HPP clinical types and pathologies, discusses early attempts at therapies for HPP, summarizes development of HPP mouse models, reviews design and validation of AA ERT, and provides up-to-date accounts of AA ERT efficacy in clinical trials and case reports, including therapeutic response, adverse effects, limitations, and potential future directions in therapy.
Collapse
Affiliation(s)
- S A Bowden
- Division of Endocrinology, Department of Pediatrics, Nationwide Children's Hospital/The Ohio State University College of Medicine, Columbus, OH, USA.
| | - B L Foster
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
28
|
Liu W, Zhang L, Xuan K, Hu C, Li L, Zhang Y, Jin F, Jin Y. Alkaline Phosphatase Controls Lineage Switching of Mesenchymal Stem Cells by Regulating the LRP6/GSK3β Complex in Hypophosphatasia. Theranostics 2018; 8:5575-5592. [PMID: 30555565 PMCID: PMC6276305 DOI: 10.7150/thno.27372] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 09/18/2018] [Indexed: 01/17/2023] Open
Abstract
Lineage differentiation of bone marrow mesenchymal stem cells (BMMSCs) is the key to bone-fat reciprocity in bone marrow. To date, the regulators of BMMSC lineage switching have all been identified to be transcription factors, and researchers have not determined whether other genes control this process. This study aims to reveal a previously unknown role of tissue-nonspecific alkaline phosphatase (TNSALP) in controlling BMMSC lineage selection. Methods: We compared the characteristics of cultured BMMSCs from patients with hypophosphatasia (HPP), which is caused by mutations in the liver/bone/kidney alkaline phosphatase (ALPL) gene, and an ALPL knockout (ko) mouse model. We performed ALPL downregulation and overexpression experiments to investigate the regulatory role of ALPL in BMMSC lineage switching. Using the PathScan array, coimmunoprecipitation experiments and pathway-guided small molecule treatments, we explored the possible mechanism underlying the regulatory effects of ALPL on cell differentiation and evaluated its therapeutic effect on ALPL ko mice. Results: BMMSCs from both patients with HPP and ALPL ko mice exhibited defective lineage differentiation, including a decrease in osteogenic differentiation and a parallel increase in adipogenic differentiation. Mechanistically, TNSALP directly interacted with LRP6 and regulated the phosphorylation of GSK3β, subsequently resulting in lineage switching of BMMSCs. Re-phosphorylation of GSK3β induced by LiCl treatment restored differentiation of BMMSCs and attenuated skeletal deformities in Alpl+/- mice. Conclusion: Based on our findings, TNSALP acts as a signal regulator to control lineage switching of BMMSCs by regulating the LRP6/GSK3β cascade.
Collapse
Affiliation(s)
- Wenjia Liu
- MS-State Key Laboratory & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, 710032, China
| | - Liqiang Zhang
- MS-State Key Laboratory & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, 710032, China
| | - Kun Xuan
- MS-State Key Laboratory & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Chenghu Hu
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, 710032, China
| | - Liya Li
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, 710032, China
| | - Yongjie Zhang
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, 710032, China
| | - Fang Jin
- MS-State Key Laboratory & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yan Jin
- MS-State Key Laboratory & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, 710032, China
| |
Collapse
|
29
|
Bowden SA, Foster BL. Profile of asfotase alfa in the treatment of hypophosphatasia: design, development, and place in therapy. Drug Des Devel Ther 2018; 12:3147-3161. [PMID: 30288020 PMCID: PMC6161731 DOI: 10.2147/dddt.s154922] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hypophosphatasia (HPP) is a multi-systemic metabolic disorder caused by loss-of-function mutations in the ALPL gene that encodes the mineralization-associated enzyme, tissue-nonspecific alkaline phosphatase (TNSALP). HPP is characterized by defective bone and dental mineralization, leading to skeletal abnormalities with complications resulting in significant morbidity and mortality. Management of HPP has been limited to supportive care until the introduction of a recently approved enzyme replacement therapy employing bone-targeted recombinant human TNSALP, asfotase alfa (AA). This new therapy has been transformative as it improves survival in severely affected infants, and overall quality of life in children and adults with HPP. This review provides an overview of HPP, focusing on important steps in the development of AA enzyme replacement therapy, including the drug design, preclinical studies in the HPP mouse model, and outcomes from clinical trials and case report publications to date, with special attention given to response to therapy of skeletal manifestations, biochemical features, and other clinical manifestations. The limitations, adverse effects, and outcomes of AA are outlined and the place in therapy for individuals with HPP is discussed.
Collapse
Affiliation(s)
- Sasigarn A Bowden
- Division of Endocrinology, Department of Pediatrics, Nationwide Children's Hospital/The Ohio State University College of Medicine, Columbus, OH 43205, USA,
| | - Brian L Foster
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH 43205, USA
| |
Collapse
|
30
|
Adam C, Glück L, Ebert R, Goebeler M, Jakob F, Schmidt M. The MEK5/ERK5 mitogen-activated protein kinase cascade is an effector pathway of bone-sustaining bisphosphonates that regulates osteogenic differentiation and mineralization. Bone 2018; 111:49-58. [PMID: 29567200 DOI: 10.1016/j.bone.2018.03.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/15/2018] [Accepted: 03/18/2018] [Indexed: 01/24/2023]
Abstract
Bisphosphonates play an important role in the treatment of metabolic bone diseases such as osteoporosis. In addition to their anti-resorptive activity by triggering osteoclast apoptosis, nitrogen-containing bisphosphonates (N-BP) may also influence osteogenic differentiation, which might rely on their capacity to inhibit the mevalonate pathway. In vascular endothelial cells inhibition of this pathway by cholesterol-lowering statins activates the MEK5/ERK5 mitogen-activated protein kinase cascade, which plays an important role in cellular differentiation, apoptosis or inflammatory processes. Here we evaluated whether N-BP may also target the MEK5/ERK5 pathway and analysed the consequences of ERK5 activation on osteogenic differentiation. We show that N-BP dose-dependently activate ERK5 in primary human endothelial cells and osteoblasts. The mechanism likely involves farnesyl pyrophosphate synthase inhibition and subsequent functional inhibition of the small GTPase Cdc42 since siRNA-mediated knockdown of both genes could reproduce N-BP-induced ERK5 activation. ERK5 activation resulted in regulation of several bone-relevant genes and was required for calcification and osteogenic differentiation of bone marrow-derived mesenchymal stems cells as evident by the lack of alkaline phosphatase induction and alizarin-red S staining observed upon ERK5 knockdown or upon differentiation initiation in presence of a pharmacological ERK5 inhibitor. Our data provide evidence that N-BP activate the MEK5/ERK5 cascade and reveal an essential role of ERK5 in osteogenic differentiation and mineralization of skeletal precursors.
Collapse
Affiliation(s)
- Christian Adam
- Department of Dermatology, University Hospital Würzburg, Germany
| | - Lucia Glück
- Department of Dermatology, University Hospital Würzburg, Germany
| | - Regina Ebert
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Germany
| | | | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Germany
| | - Marc Schmidt
- Department of Dermatology, University Hospital Würzburg, Germany.
| |
Collapse
|
31
|
Foster BL, Ao M, Salmon CR, Chavez MB, Kolli TN, Tran AB, Chu EY, Kantovitz KR, Yadav M, Narisawa S, Millán JL, Nociti FH, Somerman MJ. Osteopontin regulates dentin and alveolar bone development and mineralization. Bone 2018; 107:196-207. [PMID: 29313816 PMCID: PMC5803363 DOI: 10.1016/j.bone.2017.12.004] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 11/09/2017] [Accepted: 12/03/2017] [Indexed: 01/09/2023]
Abstract
The periodontal complex is essential for tooth attachment and function and includes the mineralized tissues, cementum and alveolar bone, separated by the unmineralized periodontal ligament (PDL). To gain insights into factors regulating cementum-PDL and bone-PDL borders and protecting against ectopic calcification within the PDL, we employed a proteomic approach to analyze PDL tissue from progressive ankylosis knock-out (Ank-/-) mice, featuring reduced PPi, rapid cementogenesis, and excessive acellular cementum. Using this approach, we identified the matrix protein osteopontin (Spp1/OPN) as an elevated factor of interest in Ank-/- mouse molar PDL. We studied the role of OPN in dental and periodontal development and function. During tooth development in wild-type (WT) mice, Spp1 mRNA was transiently expressed by cementoblasts and strongly by alveolar bone osteoblasts. Developmental analysis from 14 to 240days postnatal (dpn) indicated normal histological structures in Spp1-/- comparable to WT control mice. Microcomputed tomography (micro-CT) analysis at 30 and 90dpn revealed significantly increased volumes and tissue mineral densities of Spp1-/- mouse dentin and alveolar bone, while pulp and PDL volumes were decreased and tissue densities were increased. However, acellular cementum growth was unaltered in Spp1-/- mice. Quantitative PCR of periodontal-derived mRNA failed to identify potential local compensators influencing cementum in Spp1-/- vs. WT mice at 26dpn. We genetically deleted Spp1 on the Ank-/- mouse background to determine whether increased Spp1/OPN was regulating periodontal tissues when the PDL space is challenged by hypercementosis in Ank-/- mice. Ank-/-; Spp1-/- double deficient mice did not exhibit greater hypercementosis than that in Ank-/- mice. Based on these data, we conclude that OPN has a non-redundant role regulating formation and mineralization of dentin and bone, influences tissue properties of PDL and pulp, but does not control acellular cementum apposition. These findings may inform therapies targeted at controlling soft tissue calcification.
Collapse
Affiliation(s)
- B L Foster
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA.
| | - M Ao
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - C R Salmon
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | - 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
| | - A B Tran
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - E Y Chu
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - K R Kantovitz
- Department of Dental Materials, São Leopoldo Mandic Research Center, Campinas, São Paulo, Brazil
| | - M Yadav
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Research Institute, La Jolla, CA, USA
| | - S Narisawa
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Research Institute, La Jolla, CA, USA
| | - J L Millán
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Research Institute, La Jolla, CA, USA
| | - F H Nociti
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | - M J Somerman
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
32
|
Ao M, Chavez MB, Chu EY, Hemstreet KC, Yin Y, Yadav MC, Millán JL, Fisher LW, Goldberg HA, Somerman MJ, Foster BL. Overlapping functions of bone sialoprotein and pyrophosphate regulators in directing cementogenesis. Bone 2017; 105:134-147. [PMID: 28866368 PMCID: PMC5730356 DOI: 10.1016/j.bone.2017.08.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 12/19/2022]
Abstract
Although acellular cementum is essential for tooth attachment, factors directing its development and regeneration remain poorly understood. Inorganic pyrophosphate (PPi), a mineralization inhibitor, is a key regulator of cementum formation: tissue-nonspecific alkaline phosphatase (Alpl/TNAP) null mice (increased PPi) feature deficient cementum, while progressive ankylosis protein (Ank/ANK) null mice (decreased PPi) feature increased cementum. Bone sialoprotein (Bsp/BSP) and osteopontin (Spp1/OPN) are multifunctional extracellular matrix components of cementum proposed to have direct and indirect effects on cell activities and mineralization. Studies on dentoalveolar development of Bsp knockout (Bsp-/-) mice revealed severely reduced acellular cementum, however underlying mechanisms remain unclear. The similarity in defective cementum phenotypes between Bsp-/- mice and Alpl-/- mice (the latter featuring elevated PPi and OPN), prompted us to examine whether BSP is operating by modulating PPi-associated genes. Genetic ablation of Bsp caused a 2-fold increase in circulating PPi, altered mRNA expression of Alpl, Spp1, and Ank, and increased OPN protein in the periodontia. Generation of a Bsp knock-out (KO) cementoblast cell line revealed significantly decreased mineralization capacity, 50% increased PPi in culture media, and increased Spp1 and Ank mRNA expression. While addition of 2μg/ml recombinant BSP altered Spp1, Ank, and Enpp1 expression in cementoblasts, changes resulting from this dose were not dependent on the integrin-binding RGD motif or MAPK/ERK signaling pathway. Decreasing PPi by genetic ablation of Ank on the Bsp-/- mouse background reestablished cementum formation, allowing >3-fold increased acellular cementum volume compared to wild-type (WT). However, deleting Ank did not fully compensate for the absence of BSP. Bsp-/-; Ank-/- double-deficient mice exhibited mean 20-27% reduced cementum thickness and volume compared to Ank-/- mice. From these data, we conclude that the perturbations in PPi metabolism are not solely driving the cementum pathology in Bsp-/- mice, and that PPi is more potent than BSP as a cementum regulator, as shown by the ability to override loss of BSP by lowering PPi. We propose that BSP and PPi work in concert to direct mineralization in cementum and likely other mineralized tissues.
Collapse
Affiliation(s)
- M Ao
- 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
| | - E Y Chu
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - K C Hemstreet
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Y Yin
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - M C Yadav
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - J L Millán
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - L W Fisher
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - H A Goldberg
- Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - M J Somerman
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - B L Foster
- Biosciences Division, College of Dentistry, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
33
|
Maximiano WMA, da Silva EZM, Santana AC, de Oliveira PT, Jamur MC, Oliver C. Mast Cell Mediators Inhibit Osteoblastic Differentiation and Extracellular Matrix Mineralization. J Histochem Cytochem 2017; 65:723-741. [PMID: 28980852 DOI: 10.1369/0022155417734174] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mast cells are multifunctional immune cells that participate in many important processes such as defense against pathogens, allergic reactions, and tissue repair. These cells perform their functions through the release of a wide variety of mediators. This release occurs mainly through cross-linking IgE (immunoglobulin E) bound to high affinity IgE receptors by multivalent antigens. The abundance of mast cells in connective tissue, surrounding blood vessels, and their involvement in the early stages of bone repair support the possibility of physiological and pathological interactions between mast cells and osteoblasts. However, the participation of mast cell mediators in osteogenesis is not fully understood. Therefore, the objective of this work was to investigate the role of mast cell mediators in the acquisition of the osteogenic phenotype in vitro. The results show that pooled mast cell mediators can affect proliferation, morphology, and cytoskeleton of osteoblastic cells, and impair the activity and expression of alkaline phosphatase as well as the expression of bone sialoprotein. Also, mast cell mediators inhibit the expression of mRNA for those proteins and inhibit the formation and maturation of calcium nodules and consequently inhibit mineralization. Therefore, mast cell mediators can modulate osteogenesis and are potential therapeutic targets for treatments of bone disorders.
Collapse
Affiliation(s)
- William Marcatti Amarú Maximiano
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Elaine Zayas Marcelino da Silva
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ana Carolina Santana
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Paulo Tambasco de Oliveira
- Department of Morphology, Stomatology, and Basic Pathology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Maria Célia Jamur
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Constance Oliver
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
34
|
Halling Linder C, Ek-Rylander B, Krumpel M, Norgård M, Narisawa S, Millán JL, Andersson G, Magnusson P. Bone Alkaline Phosphatase and Tartrate-Resistant Acid Phosphatase: Potential Co-regulators of Bone Mineralization. Calcif Tissue Int 2017; 101:92-101. [PMID: 28303318 PMCID: PMC5486932 DOI: 10.1007/s00223-017-0259-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/17/2017] [Indexed: 12/18/2022]
Abstract
Phosphorylated osteopontin (OPN) inhibits hydroxyapatite crystal formation and growth, and bone alkaline phosphatase (BALP) promotes extracellular mineralization via the release of inorganic phosphate from the mineralization inhibitor inorganic pyrophosphate (PPi). Tartrate-resistant acid phosphatase (TRAP), produced by osteoclasts, osteoblasts, and osteocytes, exhibits potent phosphatase activity towards OPN; however, its potential capacity as a regulator of mineralization has not previously been addressed. We compared the efficiency of BALP and TRAP towards the endogenous substrates for BALP, i.e., PPi and pyridoxal 5'-phosphate (PLP), and their impact on mineralization in vitro via dephosphorylation of bovine milk OPN. TRAP showed higher phosphatase activity towards phosphorylated OPN and PPi compared to BALP, whereas the activity of TRAP and BALP towards PLP was comparable. Bovine milk OPN could be completely dephosphorylated by TRAP, liberating all its 28 phosphates, whereas BALP dephosphorylated at most 10 phosphates. OPN, dephosphorylated by either BALP or TRAP, showed a partially or completely attenuated phosphorylation-dependent inhibitory capacity, respectively, compared to native OPN on the formation of mineralized nodules. Thus, there are phosphorylations in OPN important for inhibition of mineralization that are removed by TRAP but not by BALP. In conclusion, our data indicate that both BALP and TRAP can alleviate the inhibitory effect of OPN on mineralization, suggesting a potential role for TRAP in skeletal mineralization. Further studies are warranted to explore the possible physiological relevance of TRAP in bone mineralization.
Collapse
Affiliation(s)
- Cecilia Halling Linder
- Department of Clinical Chemistry and Department of Clinical and Experimental Medicine, Linköping University, 581 85, Linköping, Sweden
| | - Barbro Ek-Rylander
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, 141 86, Huddinge, Sweden
| | - Michael Krumpel
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, 141 86, Huddinge, Sweden
| | - Maria Norgård
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, 141 86, Huddinge, Sweden
| | - Sonoko Narisawa
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - José Luis Millán
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Göran Andersson
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, 141 86, Huddinge, Sweden
| | - Per Magnusson
- Department of Clinical Chemistry and Department of Clinical and Experimental Medicine, Linköping University, 581 85, Linköping, Sweden.
| |
Collapse
|
35
|
Nam HK, Sharma M, Liu J, Hatch NE. Tissue Nonspecific Alkaline Phosphatase (TNAP) Regulates Cranial Base Growth and Synchondrosis Maturation. Front Physiol 2017; 8:161. [PMID: 28377728 PMCID: PMC5359511 DOI: 10.3389/fphys.2017.00161] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 03/02/2017] [Indexed: 12/27/2022] Open
Abstract
Hypophosphatasia is a rare heritable disorder caused by inactivating mutations in the gene (Alpl) that encodes tissue nonspecific alkaline phosphatase (TNAP). Hypophosphatasia with onset in infants and children can manifest as rickets. How TNAP deficiency leads to bone hypomineralization is well explained by TNAP's primary function of pyrophosphate hydrolysis when expressed in differentiated bone forming cells. How TNAP deficiency leads to abnormalities within endochondral growth plates is not yet known. Previous studies in hypophosphatemic mice showed that phosphate promotes chondrocyte maturation and apoptosis via MAPK signaling. Alpl-/- mice are not hypophosphatemic but TNAP activity does increase local levels of inorganic phosphate. Therefore, we hypothesize that TNAP influences endochondral bone development via MAPK. In support of this premise, here we demonstrate cranial base bone growth deficiency in Alpl-/- mice, utilize primary rib chondrocytes to show that TNAP influences chondrocyte maturation, apoptosis, and MAPK signaling in a cell autonomous manner; and demonstrate that similar chondrocyte signaling and apoptosis abnormalities are present in the cranial base synchondroses of Alpl-/- mice. Micro CT studies revealed diminished anterior cranial base bone and total cranial base lengths in Alpl-/- mice, that were prevented upon injection with mineral-targeted recombinant TNAP (strensiq). Histomorphometry of the inter-sphenoidal synchondrosis (cranial base growth plate) demonstrated significant expansion of the hypertrophic chondrocyte zone in Alpl-/- mice that was minimized upon treatment with recombinant TNAP. Alpl-/- primary rib chondrocytes exhibited diminished chondrocyte proliferation, aberrant mRNA expression, diminished hypertrophic chondrocyte apoptosis and diminished MAPK signaling. Diminished apoptosis and VEGF expression were also seen in 15 day-old cranial base synchondroses of Alpl-/- mice. MAPK signaling was significantly diminished in 5 day-old cranial base synchondroses of Alpl-/- mice. Together, our data suggests that TNAP is essential for the later stages of endochondral bone development including hypertrophic chondrocyte apoptosis and VEGF mediated recruitment of blood vessels for replacement of cartilage with bone. These changes may be mediated by diminished MAPK signaling in TNAP deficient chondrocytes due to diminished local inorganic phosphate production.
Collapse
Affiliation(s)
- Hwa K Nam
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan Ann Arbor, MI, USA
| | - Monika Sharma
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan Ann Arbor, MI, USA
| | - Jin Liu
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan Ann Arbor, MI, USA
| | - Nan E Hatch
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan Ann Arbor, MI, USA
| |
Collapse
|
36
|
Barron ML, Rybchyn MS, Ramesh S, Mason RS, Fiona Bonar S, Stalley P, Khosla S, Hudson B, Arthur C, Kim E, Clifton-Bligh RJ, Clifton-Bligh PB. Clinical, cellular, microscopic, and ultrastructural studies of a case of fibrogenesis imperfecta ossium. Bone Res 2017; 5:16057. [PMID: 28326223 PMCID: PMC5350113 DOI: 10.1038/boneres.2016.57] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/20/2016] [Accepted: 11/03/2016] [Indexed: 12/14/2022] Open
Abstract
Fibrogenesis imperfecta ossium is a rare disorder of bone usually characterized by marked osteopenia and associated with variable osteoporosis and osteosclerosis, changing over time. Histological examination shows that newly formed collagen is abnormal, lacking birefringence when examined by polarized light. The case presented demonstrates these features and, in addition, a previously undocumented finding of a persistent marked reduction of the serum C3 and C4. Osteoblasts established in culture from a bone biopsy showed abnormal morphology on electron microscopy and increased proliferation when cultured with benzoylbenzoyl-ATP and 1,25-dihydroxyvitamin D, contrasting with findings in normal osteoblasts in culture. A gene microarray study showed marked upregulation of the messenger RNA (mRNA) for G-protein-coupled receptor 128 (GPR 128), an orphan receptor of unknown function and also of osteoprotegerin in the patient's osteoblasts in culture. When normal osteoblasts were cultured with the patient's serum, there was marked upregulation of the mRNA for aquaporin 1. A single pathogenetic factor to account for the features of this disorder has not been defined, but the unique findings described here may facilitate more definitive investigation of the abnormal bone cell function.
Collapse
Affiliation(s)
- Melissa L Barron
- Department of Physiology, School of Medical Sciences, Bosch Institute, University of Sydney, Sydney 2006, New South Wales, Australia
| | - Mark S Rybchyn
- Department of Physiology, School of Medical Sciences, Bosch Institute, University of Sydney, Sydney 2006, New South Wales, Australia
| | - Sutharshani Ramesh
- Department of Physiology, School of Medical Sciences, Bosch Institute, University of Sydney, Sydney 2006, New South Wales, Australia
| | - Rebecca S Mason
- Department of Physiology, School of Medical Sciences, Bosch Institute, University of Sydney, Sydney 2006, New South Wales, Australia
| | - S Fiona Bonar
- Douglas HanlyMoir Pathology, Macquarie Park 2113, New South Wales, Australia
| | - Paul Stalley
- Department of Orthopaedics, Royal Prince Alfred Hospital, Camperdown 2050, New South Wales, Australia
| | - Sundeep Khosla
- Department of Endocrinology, Mayo Clinic, Rochester 55905, MN, USA
| | - Bernie Hudson
- Department of Microbiology, Royal North Shore Hospital, St Leonards 2065, New South Wales, Australia
| | - Christopher Arthur
- Department of Haematology, Royal North Shore Hospital, St Leonards 2065, New South Wales, Australia
| | - Edward Kim
- Department of Endocrinology, Royal North Shore Hospital, St Leonards 2065, New South Wales, Australia
| | - Roderick J Clifton-Bligh
- Department of Endocrinology, Royal North Shore Hospital, St Leonards 2065, New South Wales, Australia
- Faculty of Medicine, University of Sydney, Sydney 2006, New South Wales, Australia
| | - Phillip B Clifton-Bligh
- Department of Endocrinology, Royal North Shore Hospital, St Leonards 2065, New South Wales, Australia
- Faculty of Medicine, University of Sydney, Sydney 2006, New South Wales, Australia
| |
Collapse
|
37
|
Ii H, Warraich S, Tenn N, Quinonez D, Holdsworth DW, Hammond JR, Dixon SJ, Séguin CA. Disruption of biomineralization pathways in spinal tissues of a mouse model of diffuse idiopathic skeletal hyperostosis. Bone 2016; 90:37-49. [PMID: 27237608 DOI: 10.1016/j.bone.2016.05.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 05/24/2016] [Accepted: 05/25/2016] [Indexed: 01/06/2023]
Abstract
Equilibrative nucleoside transporter 1 (ENT1) mediates passage of adenosine across the plasma membrane. We reported previously that mice lacking ENT1 (ENT1(-/-)) exhibit progressive ectopic mineralization of spinal tissues resembling diffuse idiopathic skeletal hyperostosis (DISH) in humans. Here, we investigated mechanisms underlying aberrant mineralization in ENT1(-/-) mice. Micro-CT revealed ectopic mineralization of spinal tissues in both male and female ENT1(-/-) mice, involving the annulus fibrosus of the intervertebral discs (IVDs) of older mice. IVDs were isolated from wild-type and ENT1(-/-) mice at 2months of age (prior to disc mineralization), 4, and 6months of age (disc mineralization present) and processed for real-time PCR, cell isolation, or histology. Relative to the expression of ENTs in other tissues, ENT1 was the primary nucleoside transporter expressed in wild-type IVDs and mediated the functional uptake of [(3)H]2-chloroadenosine by annulus fibrosus cells. No differences in candidate gene expression were detected in IVDs from ENT1(-/-) and wild-type mice at 2 or 4months of age. However, at 6months of age, expression of genes that inhibit biomineralization Mgp, Enpp1, Ank, and Spp1 were reduced in IVDs from ENT1(-/-) mice. To assess whether changes detected in ENT1(-/-) mice were cell autonomous, annulus fibrosus cell cultures were established. Compared to wild-type cells, cells isolated from ENT1(-/-) IVDs at 2 or 6months of age demonstrated greater activity of alkaline phosphatase, a promoter of biomineralization. Cells from 2-month-old ENT1(-/-) mice also showed greater mineralization than wild-type. Interestingly, altered localization of alkaline phosphatase activity was detected in the inner annulus fibrosus of ENT1(-/-) mice in vivo. Alkaline phosphatase activity, together with the marked reduction in mineralization inhibitors, is consistent with the mineralization of IVDs seen in ENT1(-/-) mice at older ages. These findings establish that both cell-autonomous and systemic mechanisms contribute to ectopic mineralization in ENT1(-/-) mice.
Collapse
Affiliation(s)
- Hisataka Ii
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, and Bone and Joint Institute, The University of Western Ontario, London, Ontario, Canada; Department of Oral Health School of Life Dentistry at Tokyo, The Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo 102-8159, Japan
| | - Sumeeta Warraich
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, and Bone and Joint Institute, The University of Western Ontario, London, Ontario, Canada
| | - Neil Tenn
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, and Bone and Joint Institute, The University of Western Ontario, London, Ontario, Canada
| | - Diana Quinonez
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, and Bone and Joint Institute, The University of Western Ontario, London, Ontario, Canada
| | - David W Holdsworth
- Imaging Research Laboratories, Robarts Research Institute, Department of Surgery, Schulich School of Medicine & Dentistry, and Bone and Joint Institute, The University of Western Ontario, London, Ontario, Canada
| | - James R Hammond
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - S Jeffrey Dixon
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, and Bone and Joint Institute, The University of Western Ontario, London, Ontario, Canada
| | - Cheryle A Séguin
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, and Bone and Joint Institute, The University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
38
|
Okawa R, Iijima O, Kishino M, Okawa H, Toyosawa S, Sugano-Tajima H, Shimada T, Okada T, Ozono K, Ooshima T, Nakano K. Gene therapy improves dental manifestations in hypophosphatasia model mice. J Periodontal Res 2016; 52:471-478. [PMID: 27561677 DOI: 10.1111/jre.12412] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2016] [Indexed: 01/31/2023]
Abstract
BACKGROUND AND OBJECTIVE Hypophosphatasia is a rare inherited skeletal disorder characterized by defective bone mineralization and deficiency of tissue non-specific alkaline phosphatase (TNSALP) activity. The disease is caused by mutations in the liver/bone/kidney alkaline phosphatase gene (ALPL) encoding TNSALP. Early exfoliation of primary teeth owing to disturbed cementum formation, periodontal ligament weakness and alveolar bone resorption are major complications encountered in oral findings, and discovery of early loss of primary teeth in a dental examination often leads to early diagnosis of hypophosphatasia. Although there are no known fundamental treatments or effective dental approaches to prevent early exfoliation of primary teeth in affected patients, several possible treatments have recently been described, including gene therapy. Gene therapy has also been applied to TNSALP knockout mice (Alpl-/- ), which phenocopy the infantile form of hypophosphatasia, and improved their systemic condition. In the present study, we investigated whether gene therapy improved the dental condition of Alpl-/- mice. MATERIAL AND METHODS Following sublethal irradiation (4 Gy) at the age of 2 d, Alpl-/- mice underwent gene therapy using bone marrow cells transduced with a lentiviral vector expressing a bone-targeted form of TNSALP injected into the jugular vein (n = 3). Wild-type (Alpl+/+ ), heterozygous mice (Alpl+/- ) and Alpl-/- mice were analyzed at 9 d of age (n = 3 of each), while Alpl+/+ mice and treated or untreated Alpl-/- mice were analyzed at 1 mo of age (n = 3 of each), and Alpl+/- mice and Alpl-/- mice with gene therapy were analyzed at 3 mo of age (n = 3 of each). A single mandibular hemi-section obtained at 1 mo of age was analyzed using a small animal computed tomography machine to assess alveolar bone formation. Other mandibular hemi-sections obtained at 9 d, 1 mo and 3 mo of age were subjected to hematoxylin and eosin staining and immunohistochemical analysis of osteopontin, a marker of cementum. RESULTS Immunohistochemical analysis of osteopontin, a marker of acellular cementum, revealed that Alpl-/- mice displayed impaired formation of cementum and alveolar bone, similar to the human dental phenotype. Cementum formation was clearly present in Alpl-/- mice that underwent gene therapy, but did not recover to the same level as that in wild-type (Alpl+/+ ) mice. Micro-computed tomography examination showed that gene therapy improved alveolar bone mineral density in Alpl-/- mice to a similar level to that in Alpl+/+ mice. CONCLUSIONS Our results suggest that gene therapy can improve the general condition of Alpl-/- mice, and induce significant alveolar bone formation and moderate improvement of cementum formation, which may contribute to inhibition of early spontaneous tooth exfoliation.
Collapse
Affiliation(s)
- R Okawa
- Osaka University Graduate School of Dentistry, Osaka, Japan
| | - O Iijima
- Nippon Medical School, Tokyo, Japan
| | - M Kishino
- Osaka University Graduate School of Dentistry, Osaka, Japan
| | - H Okawa
- Osaka University Graduate School of Dentistry, Osaka, Japan
| | - S Toyosawa
- Osaka University Graduate School of Dentistry, Osaka, Japan
| | | | | | - T Okada
- Nippon Medical School, Tokyo, Japan
| | - K Ozono
- Osaka University Graduate School of Medicine, Osaka, Japan
| | - T Ooshima
- Osaka University Graduate School of Dentistry, Osaka, Japan
| | - K Nakano
- Osaka University Graduate School of Dentistry, Osaka, Japan
| |
Collapse
|
39
|
Yadav MC, Bottini M, Cory E, Bhattacharya K, Kuss P, Narisawa S, Sah RL, Beck L, Fadeel B, Farquharson C, Millán JL. Skeletal Mineralization Deficits and Impaired Biogenesis and Function of Chondrocyte-Derived Matrix Vesicles in Phospho1(-/-) and Phospho1/Pi t1 Double-Knockout Mice. J Bone Miner Res 2016; 31:1275-86. [PMID: 26773408 PMCID: PMC4891278 DOI: 10.1002/jbmr.2790] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 01/05/2016] [Accepted: 01/13/2016] [Indexed: 11/10/2022]
Abstract
We have previously shown that ablation of either the Phospho1 or Alpl gene, encoding PHOSPHO1 and tissue-nonspecific alkaline phosphatase (TNAP) respectively, lead to hyperosteoidosis, but that their chondrocyte-derived and osteoblast-derived matrix vesicles (MVs) are able to initiate mineralization. In contrast, the double ablation of Phospho1 and Alpl completely abolish initiation and progression of skeletal mineralization. We argued that MVs initiate mineralization by a dual mechanism: PHOSPHO1-mediated intravesicular generation of inorganic phosphate (Pi ) and phosphate transporter-mediated influx of Pi . To test this hypothesis, we generated mice with col2a1-driven Cre-mediated ablation of Slc20a1, hereafter referred to as Pi t1, alone or in combination with a Phospho1 gene deletion. Pi t1(col2/col2) mice did not show any major phenotypic abnormalities, whereas severe skeletal deformities were observed in the [Phospho1(-/-) ; Pi t1(col2/col2) ] double knockout mice that were more pronounced than those observed in the Phospho1(-/-) mice. Histological analysis of [Phospho1(-/-) ; Pi t1(col2/col2) ] bones showed growth plate abnormalities with a shorter hypertrophic chondrocyte zone and extensive hyperosteoidosis. The [Phospho1(-/-) ; Pi t1(col2/col2) ] skeleton displayed significant decreases in BV/TV%, trabecular number, and bone mineral density, as well as decreased stiffness, decreased strength, and increased postyield deflection compared to Phospho1(-/-) mice. Using atomic force microscopy we found that ∼80% of [Phospho1(-/-) ; Pi t1(col2/col2) ] MVs were devoid of mineral in comparison to ∼50% for the Phospho1(-/-) MVs and ∼25% for the WT and Pi t1(col2/col2) MVs. We also found a significant decrease in the number of MVs produced by both Phospho1(-/-) and [Phospho1(-/-) ; Pi t1(col2/col2) ] chondrocytes. These data support the involvement of phosphate transporter 1, hereafter referred to as Pi T-1, in the initiation of skeletal mineralization and provide compelling evidence that PHOSPHO1 function is involved in MV biogenesis. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Manisha C Yadav
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Massimo Bottini
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy.,Inflammatory and Infectious Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Esther Cory
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Kunal Bhattacharya
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Pia Kuss
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Sonoko Narisawa
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Robert L Sah
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Laurent Beck
- INSERM U791, Centre for Osteoarticular and Dental Tissue Engineering (LIOAD), Nantes, Cedex, France
| | - Bengt Fadeel
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Colin Farquharson
- The Roslin Institute, The University of Edinburgh, Midlothian, Scotland, UK
| | - José Luis Millán
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
40
|
Abstract
Hypophosphatasia (HPP) results from ALPL mutations leading to deficient activity of the tissue-non-specific alkaline phosphatase isozyme (TNAP) and thereby extracellular accumulation of inorganic pyrophosphate (PPi), a natural substrate of TNAP and potent inhibitor of mineralization. Thus, HPP features rickets or osteomalacia and hypomineralization of teeth. Enzyme replacement using mineral-targeted TNAP from birth prevented severe HPP in TNAP-knockout mice and was then shown to rescue and substantially treat infants and young children with life-threatening HPP. Clinical trials are revealing aspects of HPP pathophysiology not yet fully understood, such as craniosynostosis and muscle weakness when HPP is severe. New treatment approaches are under development to improve patient care.
Collapse
Affiliation(s)
- José Luis Millán
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA.
| | - Michael P Whyte
- Center for Metabolic Bone Disease and Molecular Research, Shriners Hospital for Children, St. Louis, MO, 63110, USA
- Division of Bone and Mineral Diseases, Washington University School of Medicine at Barnes-Jewish Hospital, St. Louis, MO, 63110, USA
| |
Collapse
|
41
|
Bouleftour W, Juignet L, Bouet G, Granito RN, Vanden-Bossche A, Laroche N, Aubin JE, Lafage-Proust MH, Vico L, Malaval L. The role of the SIBLING, Bone Sialoprotein in skeletal biology - Contribution of mouse experimental genetics. Matrix Biol 2016; 52-54:60-77. [PMID: 26763578 DOI: 10.1016/j.matbio.2015.12.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/23/2015] [Accepted: 12/28/2015] [Indexed: 12/17/2022]
Abstract
Bone Sialoprotein (BSP) is a member of the "Small Integrin-Binding Ligand N-linked Glycoproteins" (SIBLING) extracellular matrix protein family of mineralized tissues. BSP has been less studied than other SIBLING proteins such as Osteopontin (OPN), which is coexpressed with it in several skeletal cell types. Here we review the contribution of genetically engineered mice (BSP gene knockout and overexpression) to the understanding of the role of BSP in the bone organ. The studies made so far highlight the role of BSP in skeletal mineralization, as well as its importance for proper osteoblast and osteoclast differentiation and activity, most prominently in primary/repair bone. The absence of BSP also affects the local environment of the bone tissue, in particular hematopoiesis and vascularization. Interestingly, lack of BSP induces an overexpression of OPN, and the cognate protein could be responsible for some aspects of the BSP gene knockout skeletal phenotype, while replacing BSP for some of its functions. Such interplay between the partly overlapping functions of SIBLING proteins, as well as the network of cross-regulations in which they are involved should now be the focus of further work.
Collapse
Affiliation(s)
- Wafa Bouleftour
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Laura Juignet
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Guenaelle Bouet
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge, UK
| | | | - Arnaud Vanden-Bossche
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Norbert Laroche
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Jane E Aubin
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Marie-Hélène Lafage-Proust
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Laurence Vico
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France
| | - Luc Malaval
- Université de Lyon - Université Jean Monnet, INSERM U1059-LBTO/IFRESIS, Faculté de Médecine, 10 Chemin de la Marandière, St Priest en Jarez F42270, France.
| |
Collapse
|
42
|
Javaheri B, Carriero A, Staines KA, Chang YM, Houston DA, Oldknow KJ, Millan JL, Kazeruni BN, Salmon P, Shefelbine S, Farquharson C, Pitsillides AA. Phospho1 deficiency transiently modifies bone architecture yet produces consistent modification in osteocyte differentiation and vascular porosity with ageing. Bone 2015; 81:277-291. [PMID: 26232374 PMCID: PMC4652607 DOI: 10.1016/j.bone.2015.07.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 07/24/2015] [Accepted: 07/27/2015] [Indexed: 12/11/2022]
Abstract
PHOSPHO1 is one of principal proteins involved in initiating bone matrix mineralisation. Recent studies have found that Phospho1 KO mice (Phospho1-R74X) display multiple skeletal abnormalities with spontaneous fractures, bowed long bones, osteomalacia and scoliosis. These analyses have however been limited to young mice and it remains unclear whether the role of PHOSPHO1 is conserved in the mature murine skeleton where bone turnover is limited. In this study, we have used ex-vivo computerised tomography to examine the effect of Phospho1 deletion on tibial bone architecture in mice at a range of ages (5, 7, 16 and 34 weeks of age) to establish whether its role is conserved during skeletal growth and maturation. Matrix mineralisation has also been reported to influence terminal osteoblast differentiation into osteocytes and we have also explored whether hypomineralised bones in Phospho1 KO mice exhibit modified osteocyte lacunar and vascular porosity. Our data reveal that Phospho1 deficiency generates age-related defects in trabecular architecture and compromised cortical microarchitecture with greater porosity accompanied by marked alterations in osteocyte shape, significant increases in osteocytic lacuna and vessel number. Our in vitro studies examining the behaviour of osteoblast derived from Phospho1 KO and wild-type mice reveal reduced levels of matrix mineralisation and modified osteocytogenic programming in cells deficient in PHOSPHO1. Together our data suggest that deficiency in PHOSPHO1 exerts modifications in bone architecture that are transient and depend upon age, yet produces consistent modification in lacunar and vascular porosity. It is possible that the inhibitory role of PHOSPHO1 on osteocyte differentiation leads to these age-related changes in bone architecture. It is also intriguing to note that this apparent acceleration in osteocyte differentiation evident in the hypomineralised bones of Phospho1 KO mice suggests an uncoupling of the interplay between osteocytogenesis and biomineralisation. Further studies are required to dissect the molecular processes underlying the regulatory influences exerted by PHOSPHO1 on the skeleton with ageing.
Collapse
Affiliation(s)
- B Javaheri
- The Royal Veterinary College, London, United Kingdom.
| | - A Carriero
- Department of Biomedical Engineering, Florida Institute of Technology Melbourne, FL 32901, USA
| | - K A Staines
- The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, United Kingdom
| | - Y-M Chang
- The Royal Veterinary College, London, United Kingdom
| | - D A Houston
- The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, United Kingdom
| | - K J Oldknow
- The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, United Kingdom
| | - J L Millan
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | | | - P Salmon
- Bruker-microCT, Kartuizersweg 3B, 2550 Kontich, Belgium
| | - S Shefelbine
- Department of Mechanical and Industrial Engineering, Northeastern University, USA
| | - C Farquharson
- The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, United Kingdom
| | | |
Collapse
|
43
|
Liu J, Campbell C, Nam HK, Caron A, Yadav MC, Millán JL, Hatch NE. Enzyme replacement for craniofacial skeletal defects and craniosynostosis in murine hypophosphatasia. Bone 2015; 78:203-11. [PMID: 25959417 PMCID: PMC4466206 DOI: 10.1016/j.bone.2015.05.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 04/13/2015] [Accepted: 05/02/2015] [Indexed: 12/30/2022]
Abstract
Hypophosphatasia (HPP) is an inborn-error-of-metabolism disorder characterized by deficient bone and tooth mineralization due to loss-of function mutations in the gene (Alpl) encoding tissue-nonspecific alkaline phosphatase (TNAP). Alpl(-/-) mice exhibit many characteristics seen in infantile HPP including long bone and tooth defects, vitamin B6 responsive seizures and craniosynostosis. Previous reports demonstrated that a mineral-targeted form of TNAP rescues long bone, vertebral and tooth mineralization defects in Alpl(-/-) mice. Here we report that enzyme replacement with mineral-targeted TNAP (asfotase-alfa) also prevents craniosynostosis (the premature fusion of cranial bones) and additional craniofacial skeletal abnormalities in Alpl(-/-) mice. Craniosynostosis, cranial bone volume and density, and craniofacial shape abnormalities were assessed by microscopy, histology, digital caliper measurements and micro CT. We found that craniofacial shape defects, cranial bone mineralization and craniosynostosis were corrected in Alpl(-/-) mice injected daily subcutaneously starting at birth with recombinant enzyme. Analysis of Alpl(-/-) calvarial cells indicates that TNAP deficiency leads to aberrant osteoblastic gene expression and diminished proliferation. Some but not all of these cellular abnormalities were rescued by treatment with inorganic phosphate. These results confirm an essential role for TNAP in craniofacial skeletal development and demonstrate the efficacy of early postnatal mineral-targeted enzyme replacement for preventing craniofacial abnormalities including craniosynostosis in murine infantile HPP.
Collapse
Affiliation(s)
- Jin Liu
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Cassie Campbell
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Hwa Kyung Nam
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | | | - Manisha C Yadav
- Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | | | - Nan E Hatch
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA..
| |
Collapse
|
44
|
Komarova SV, Safranek L, Gopalakrishnan J, Ou MJY, McKee MD, Murshed M, Rauch F, Zuhr E. Mathematical model for bone mineralization. Front Cell Dev Biol 2015; 3:51. [PMID: 26347868 PMCID: PMC4544393 DOI: 10.3389/fcell.2015.00051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/04/2015] [Indexed: 12/23/2022] Open
Abstract
Defective bone mineralization has serious clinical manifestations, including deformities and fractures, but the regulation of this extracellular process is not fully understood. We have developed a mathematical model consisting of ordinary differential equations that describe collagen maturation, production and degradation of inhibitors, and mineral nucleation and growth. We examined the roles of individual processes in generating normal and abnormal mineralization patterns characterized using two outcome measures: mineralization lag time and degree of mineralization. Model parameters describing the formation of hydroxyapatite mineral on the nucleating centers most potently affected the degree of mineralization, while the parameters describing inhibitor homeostasis most effectively changed the mineralization lag time. Of interest, a parameter describing the rate of matrix maturation emerged as being capable of counter-intuitively increasing both the mineralization lag time and the degree of mineralization. We validated the accuracy of model predictions using known diseases of bone mineralization such as osteogenesis imperfecta and X-linked hypophosphatemia. The model successfully describes the highly nonlinear mineralization dynamics, which includes an initial lag phase when osteoid is present but no mineralization is evident, then fast primary mineralization, followed by secondary mineralization characterized by a continuous slow increase in bone mineral content. The developed model can potentially predict the function for a mutated protein based on the histology of pathologic bone samples from mineralization disorders of unknown etiology.
Collapse
Affiliation(s)
- Svetlana V Komarova
- Faculty of Dentistry, McGill University Montreal, QC, Canada ; Shriners Hospital for Children-Canada Montreal, QC, Canada
| | - Lee Safranek
- Department of Mathematics, Simon Fraser University Burnaby, BC, Canada
| | - Jay Gopalakrishnan
- The Fariborz Maseeh Department of Mathematics and Statistics, Portland State University Portland, OR, USA
| | | | - Marc D McKee
- Faculty of Dentistry, McGill University Montreal, QC, Canada ; Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University Montreal, QC, Canada
| | - Monzur Murshed
- Faculty of Dentistry, McGill University Montreal, QC, Canada ; Shriners Hospital for Children-Canada Montreal, QC, Canada ; Department of Medicine, Faculty of Medicine, McGill University Montreal, QC, Canada
| | - Frank Rauch
- Shriners Hospital for Children-Canada Montreal, QC, Canada
| | - Erica Zuhr
- Department of Mathematics, High Point University High Point, NC, USA
| |
Collapse
|
45
|
Lee JW, Park JW, Khang D. Analysis of Osteoblast Differentiation on Polymer Thin Films Embedded with Carbon Nanotubes. PLoS One 2015; 10:e0129856. [PMID: 26076355 PMCID: PMC4468207 DOI: 10.1371/journal.pone.0129856] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/12/2015] [Indexed: 11/18/2022] Open
Abstract
Osteoblast differentiation can be modulated by variations in order of nanoscale topography. Biopolymers embedded with carbon nanotubes can cause various orders of roughness at the nanoscale and can be used to investigate the dynamics of extracellular matrix interaction with cells. In this study, clear relationship between the response of osteoblasts to integrin receptor activation, their phenotype, and transcription of certain genes on polymer composites embedded with carbon nanotubes was demonstrated. We generated an ultrathin nanocomposite film embedded with carbon nanotubes and observed improved adhesion of pre-osteoblasts, with a subsequent increase in their proliferation. The expression of genes encoding integrin subunits α5, αv, β1, and β3 was significantly upregulated at the early of time-point when cells initially attached to the carbon nanotube/polymer composite. The advantage of ultrathin nanocomposite film for pre-osteoblasts was demonstrated by staining for the cytoskeletal protein vinculin and cell nuclei. The expression of essential transcription factors for osteoblastogenesis, such as Runx2 and Sp7 transcription factor 7 (known as osterix), was upregulated after 7 days. Consequently, the expression of genes that determine osteoblast phenotype, such as alkaline phosphatase, type I collagen, and osteocalcin, was accelerated on carbon nanotube embedded polymer matrix after 14 days. In conclusion, the ultrathin nanocomposite film generated various orders of nanoscale topography that triggered processes related to osteoblast bone formation.
Collapse
Affiliation(s)
- Jin Woo Lee
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, South Korea
| | - Jin-Woo Park
- Department of Periodontology, School of Dentistry, Kyungpook National University, Daegu, South Korea
- * E-mail: (J-WP); (DK)
| | - Dongwoo Khang
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, South Korea
- * E-mail: (J-WP); (DK)
| |
Collapse
|
46
|
The role of fetuin-A in mineral trafficking and deposition. BONEKEY REPORTS 2015; 4:672. [PMID: 25987986 DOI: 10.1038/bonekey.2015.39] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/13/2015] [Indexed: 12/18/2022]
Abstract
Calcium and phosphate are the principle ions involved in the deposition of mineral in the human body. Inhibitors of mineralisation are essential for the prevention of ectopic mineral precipitation and deposition. In the past decade, through in vitro, in vivo and clinical observation studies, we have come to appreciate the importance of fetuin-A (Fet-A), a circulating glycoprotein, in preventing ectopic calcium phosphate mineralisation. Moreover, the detection of Fet-A-containing mineral complex, termed calciprotein particles (CPPs), has provided new ways to assess an individual's calcific risk. The pathophysiological significance of CPPs in disease states is yet to be defined, but it provides an exciting avenue to further our understanding of the development of ectopic mineralisation.
Collapse
|
47
|
Foster BL, Sheen CR, Hatch NE, Liu J, Cory E, Narisawa S, Kiffer-Moreira T, Sah RL, Whyte MP, Somerman MJ, Millán JL. Periodontal Defects in the A116T Knock-in Murine Model of Odontohypophosphatasia. J Dent Res 2015; 94:706-14. [PMID: 25716980 DOI: 10.1177/0022034515573273] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mutations in ALPL result in hypophosphatasia (HPP), a disease causing defective skeletal mineralization. ALPL encodes tissue nonspecific alkaline phosphatase (ALP), an enzyme that promotes mineralization by reducing inorganic pyrophosphate, a mineralization inhibitor. In addition to skeletal defects, HPP causes dental defects, and a mild clinical form of HPP, odontohypophosphatasia, features only a dental phenotype. The Alpl knockout (Alpl (-/-)) mouse phenocopies severe infantile HPP, including profound skeletal and dental defects. However, the severity of disease in Alpl (-/-) mice prevents analysis at advanced ages, including studies to target rescue of dental tissues. We aimed to generate a knock-in mouse model of odontohypophosphatasia with a primarily dental phenotype, based on a mutation (c.346G>A) identified in a human kindred with autosomal dominant odontohypophosphatasia. Biochemical, skeletal, and dental analyses were performed on the resulting Alpl(+/A116T) mice to validate this model. Alpl(+/A116T) mice featured 50% reduction in plasma ALP activity compared with wild-type controls. No differences in litter size, survival, or body weight were observed in Alpl(+/A116T) versus wild-type mice. The postcranial skeleton of Alpl(+/A116T) mice was normal by radiography, with no differences in femur length, cortical/trabecular structure or mineral density, or mechanical properties. Parietal bone trabecular compartment was mildly altered. Alpl(+/A116T) mice featured alterations in the alveolar bone, including radiolucencies and resorptive lesions, osteoid accumulation on the alveolar bone crest, and significant differences in several bone properties measured by micro-computed tomography. Nonsignificant changes in acellular cementum did not appear to affect periodontal attachment or function, although circulating ALP activity was correlated significantly with incisor cementum thickness. The Alpl(+/A116T) mouse is the first model of odontohypophosphatasia, providing insights on dentoalveolar development and function under reduced ALP, bringing attention to direct effects of HPP on alveolar bone, and offering a new model for testing potential dental-targeted therapies in future studies.
Collapse
Affiliation(s)
- B L Foster
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - C R Sheen
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - N E Hatch
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - J Liu
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - E Cory
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - S Narisawa
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - T Kiffer-Moreira
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - R L Sah
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - M P Whyte
- Center for Metabolic Bone Disease and Molecular Research, Shriners Hospital for Children, St. Louis, MO, USA Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - M J Somerman
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - J L Millán
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| |
Collapse
|
48
|
Abstract
Biomineralization is a complex process in the development of mineralized tissues such as bone and pathological calcifications such as atherosclerotic plaques, kidney stones and gout. Osteopontin (OPN), an anionic phosphoprotein, is expressed in mineralizing tissues and has previously been demonstrated to be a potent inhibitor of hydroxyapatite formation. The OPN-deficient (Opn-/-) mouse displays a hypermineralized bone phenotype starting at 12 weeks postnatally. By isolating and culturing Opn-/- and wild-type (WT) osteoblasts, we sought to determine the role of OPN and two of its functional peptides in osteoblast development and mineralization. Opn-/- osteoblasts had significantly increased mineral deposition relative to their WT counterparts, with no physiologically relevant change in gene expression of osteogenic markers. Supplementation with bovine milk OPN (mOPN) led to a dramatic reduction in mineral deposition by the Opn-/- osteoblasts. Treatment with OPN-derived peptides corresponding to phosphorylated OPN-(220-235) (P3) and non-phosphorylated OPN-(65-80) (OPAR) also rescued the hypermineralization phenotype of Opn-/- osteogenic cultures. Supplementation with mOPN or the OPN-derived peptides did not alter the expression of terminal osteogenic markers. These data suggest that OPN plays an important role in the regulation of biomineralization, but that OPN does not appear to affect osteoblast cell development in vitro.
Collapse
|
49
|
Abstract
Hypophosphatasia (HPP) is due to mutations of the tissue non-specific alkaline phosphatase (TNAP) gene expressed in the liver, kidney, and bone. TNAP substrates include inorganic pyrophosphate cleaved into inorganic phosphate (Pi) in bone, pyridoxal-5'-phosphate (PLP), the circulating form of vitamin B6, and phosphoethanolamine (PEA). As an autosomal recessive or dominant disease, HPP results in a range of clinical forms. Its hallmarks are low alkaline phosphatase (AP) and elevated PLP and PEA levels. Perinatal HPP may cause early death with respiratory insufficiency and hypomineralization resulting in deformed limbs and sometimes near-absence of bones and skull. Infantile HPP is diagnosed before 6 months of life. Respiratory failure, rib fractures and seizures due to vitamin B6 deficiency in the brain indicate poor prognosis. Craniosynostosis is frequent. Unlike in other forms of rickets, calcium and phosphorus are not decreased, resulting in hypercalciuria and nephrocalcinosis. Hypercalcemic crisis may occur. Failure to thrive and growth retardation are concerns. In infantile and adult forms of HPP, non-traumatic fractures may be the prominent manifestation, with otherwise unexplained chronic pain. Progressive myopathy has been described. Dental manifestations with early loss of teeth are usual in HPP and in a specific form, odontohypophosphatasia. HPP has been studied in knock-out mice models which mimic its severe form. Animal models have made a major contribution to the development of an original enzyme therapy for human infantile HPP, which is however essentially targeted at mineralized tissues. Better knowledge of its extraskeletal manifestations, including pain and neurological symptoms, is therefore required.
Collapse
Affiliation(s)
- Jean Pierre Salles
- Unité d'Endocrinologie Maladies Osseuses, Hôpital Des Enfants, CHU de Toulouse and Inserm UMR 1043 UPS, Toulouse Cedex, France,
| |
Collapse
|
50
|
Abstract
Genetically modified mice are powerful tools for understanding the functions of genes and proteins and often serve as models of human disease. Here, several knockout and transgenic mouse lines related to tissue-nonspecific alkaline phosphatase (TNAP) are described. Conventional TNAP knockout mice die before weaning and show vitamin B6 dependent epilepsy and impaired bone mineralization, mimicking infantile hypophosphatasia. Administration of recombinant human TNAP rescues the lethal phenotype and improves bone mineralization in the null knockout mice, and this enzyme replacement therapy has been successfully applied to the treatment of human patients. Transgenic expression of human TNAP also rescues the TNAP knockout mice. Studies of the TNAP knockout mice and their double knockouts with ectonucleotide pyrophosphatase/phosphodiesterase 1 or progressive ankylosis protein revealed that pyridoxal phosphate and inorganic pyrophosphate are natural substrates of TNAP. Bone osteopontin from TNAP knockout mice is highly phosphorylated, whereas osteopontin from TNAP knockout mice expressing human TNAP is de-phosphorylated, similar to that in wild type mice, indicating that osteopontin is also a natural substrate of TNAP and that phosphorylated osteopontin contributes the impaired bone mineralization in TNAP knockout mice. Conditional TNAP knockout mice and TNAP mutants produced by ENU (N-ethyl-N-nitrosourea) mutagenesis show milder hypophosphatasia and are expected to be useful models of adult hypophosphatasia.
Collapse
|