1
|
Akitomo T, Niizato N, Kaneki A, Ogawa M, Nishimura T, Kametani M, Usuda M, Iwamoto Y, Mitsuhata C, Nomura R. A Case of Hypophosphatasia Started Enzyme Replacement Therapy Since Babyhood Stage. CHILDREN (BASEL, SWITZERLAND) 2025; 12:61. [PMID: 39857892 PMCID: PMC11764115 DOI: 10.3390/children12010061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Hypophosphatasia (HPP) is an inherited disease caused by low activity of tissue-nonspecific alkaline phosphatase. Dental characteristics include premature loss of primary teeth, enlarged pulp chambers, and enamel hypoplasia. Although enzyme replacement therapy with asfotase alfa was approved in 2015, there are few reports about the dental outcomes of this treatment. CASE PRESENTATION A 1-year-old girl referred to our hospital had already lost two primary teeth at the time of her initial visit. She started enzyme replacement therapy 6 days after birth, and genetic analysis later confirmed the diagnosis of HPP. At the age of 4 years and 7 months, 11 primary teeth had been lost, and some of the exfoliated teeth showed inflammatory root resorption or root fracture. There was also a history of abscess formation in a non-carious primary molar. CONCLUSIONS This report suggests that early enzyme replacement therapy may prevent traditional tooth loss in patients with HPP. It also highlights the new challenges posed for dental professionals in providing infection control in large pulp cavities and receding periodontal tissue.
Collapse
Affiliation(s)
- Tatsuya Akitomo
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan; (A.K.); (M.O.); (T.N.); (M.K.); (M.U.); (Y.I.); (C.M.); (R.N.)
| | | | - Ami Kaneki
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan; (A.K.); (M.O.); (T.N.); (M.K.); (M.U.); (Y.I.); (C.M.); (R.N.)
| | - Masashi Ogawa
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan; (A.K.); (M.O.); (T.N.); (M.K.); (M.U.); (Y.I.); (C.M.); (R.N.)
| | - Taku Nishimura
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan; (A.K.); (M.O.); (T.N.); (M.K.); (M.U.); (Y.I.); (C.M.); (R.N.)
| | - Mariko Kametani
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan; (A.K.); (M.O.); (T.N.); (M.K.); (M.U.); (Y.I.); (C.M.); (R.N.)
| | - Momoko Usuda
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan; (A.K.); (M.O.); (T.N.); (M.K.); (M.U.); (Y.I.); (C.M.); (R.N.)
| | - Yuko Iwamoto
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan; (A.K.); (M.O.); (T.N.); (M.K.); (M.U.); (Y.I.); (C.M.); (R.N.)
| | - Chieko Mitsuhata
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan; (A.K.); (M.O.); (T.N.); (M.K.); (M.U.); (Y.I.); (C.M.); (R.N.)
| | - Ryota Nomura
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan; (A.K.); (M.O.); (T.N.); (M.K.); (M.U.); (Y.I.); (C.M.); (R.N.)
| |
Collapse
|
2
|
Shirinezhad A, Esmaeili S, Azarboo A, Tavakoli Y, Hoveidaei AH, Zareshahi N, Ghaseminejad-Raeini A. Efficacy and safety of asfotase alfa in patients with hypophosphatasia: A systematic review. Bone 2024; 188:117219. [PMID: 39089608 DOI: 10.1016/j.bone.2024.117219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/18/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Hypophosphatasia (HPP) is a rare genetic disorder characterized by defective bone mineralization, leading to skeletal abnormalities and systemic complications. Asfotase alfa, a recombinant human tissue-nonspecific alkaline phosphatase (TNSALP) enzyme replacement therapy, has emerged as a promising treatment for HPP. However, a comprehensive evaluation of its efficacy and safety is warranted to guide clinical practice effectively. METHODS The study followed Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines and was registered in Prospective Register of Systematic Reviews (PROSPERO). A search strategy across databases found studies on asfotase alfa for HPP. Two researchers independently extracted and assessed data. This systematic review examined how the drug impacted clinical outcomes such as survival rates, musculoskeletal symptoms, respiratory function, growth measurements, dental health, quality of life, and laboratory results. RESULTS This systematic review included 15 articles with a total of 455 HPP patients. Asfotase alfa was predominantly administered at a dose of 6 mg per kg per week among the reviewed studies. Notable findings included enhanced survival rates, relief from musculoskeletal pain, improvements in respiratory outcomes, growth parameters, dental health, and quality of life. Changes in laboratory variables indicated positive responses to treatment, including changes such as increase in alkaline phosphatase (ALP), decline in pyridoxal 5'-phosphate (PLP) and inorganic pyrophosphate (PPi) levels. CONCLUSION Asfotase alfa demonstrates efficacy in improving clinical outcomes and safety in patients with HPP. Its therapeutic benefits extend across various domains. However, Larger, age-stratified comparative studies are needed to further investigate the drug's effects in HPP patients.
Collapse
Affiliation(s)
| | - Sina Esmaeili
- Sina University Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Azarboo
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Yasaman Tavakoli
- Student Research Committee, Department of Medicine, Mazandaran University of Medical Sciences, Mazandaran, Iran
| | - Amir Human Hoveidaei
- International Center for Limb Lengthening, Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, Maryland, USA
| | - Negar Zareshahi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
3
|
Chen Y, Hao Y, Chen J, Han Q, Wang Z, Peng X, Cheng L. Lacticaseibacillus rhamnosus inhibits the development of dental caries in rat caries model and in vitro. J Dent 2024; 149:105278. [PMID: 39111536 DOI: 10.1016/j.jdent.2024.105278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024] Open
Abstract
OBJECTIVES Dental caries result from a microbial imbalance in the oral cavity. Probiotics ecologically modulate the oral microflora to prevent caries. This study evaluated the anti-cariogenic effects of two Lacticaseibacillus rhamnosus strains in vitro and in vivo to provide a more theoretical basis for its clinical applications in caries prevention. METHODS In the study, cariogenic biofilms were grown with L. rhamnosus (LGG) or L. rhamnosus ATCC 7469 and analyzed. Quantitative real-time PCR (qPCR), Scanning Electron Microscope (SEM), and Confocal laser scanning microscope (CLSM) were used to detect the changes in the composition and architectures; cariogenic activity was measured by the lactic acid production and Transverse Microradiography (TMR). The effects of LGG on the 12 Sprague-Dawley rat caries model were assessed using Keyes scores and micro-CT analysis. Oral microbiome changes were evaluated through 16S rRNA gene high-throughput sequencing. RESULTS L. rhamnosus can reduce cariogenic bacteria in biofilm by 14.7 % to 48.9 %, with LGG exhibiting more potent inhibitory effects. Both strains of L. rhamnosus can adhere to the surface of biofilms, reduce the extracellular polysaccharides (EPS) matrix, and loosen the biofilm structure. L. rhamnosus inhibited cariogenic activity by reducing the lactic acid production in biofilms. The bovine enamel blocks presented lower mineral loss values and lesion depth values in the group Core+L.rh and Core+LGG. LGG-ingested rats had significantly lower levels of moderate dentin lesions and higher mineral density than the control group. The 16 s rRNA gene sequencing revealed that LGG regulated the beta diversity of the oral microbial community in the rat dental caries model. CONCLUSIONS This study revealed the promising potential of L. rhamnosus, especially the LGG strain, in the ecological prevention of dental caries. CLINICAL SIGNIFICANCE Probiotics may provide a strategy for preventing caries by regulating the oral microecological balance. The study revealed the promising anti-caries potential of the LGG probiotic strain in vivo and in vitro. It is expected that LGG could be used as an oral probiotic for the clinical prevention and treatment of caries.
Collapse
Affiliation(s)
- Yanyan Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontic, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China; Department of Stomatology, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Yu Hao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontic, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jing Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontic, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Qi Han
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontic, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zheng Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontic, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xian Peng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontic, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Operative Dentistry and Endodontic, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
4
|
Nagasaki K, Nagasaki A, Taylor JM, Kear BD, Ma Y, Somerman MJ, Gavrilova O. The RGD region of bone sialoprotein affects metabolic activity in mice. FRONTIERS IN DENTAL MEDICINE 2023; 4:1124084. [PMID: 39916931 PMCID: PMC11797800 DOI: 10.3389/fdmed.2023.1124084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/23/2023] [Indexed: 02/09/2025] Open
Abstract
Introduction Bone sialoprotein (BSP) is a key regulator of mineralized tissue formation. Previously, we generated BSP-KAE knock-in mice (KAEKI mice) by substituting a non-function KAE (lysine-alanine-glutamic acid) for the integrin-binding RGD (arginine-glycine-aspartic acid) sequence and reported a vital role of the BSP-RGD motif in modulating the periodontal ligament (PDL). Specifically, a histological disorganization of the PDL was noted, resulting in a weakened function of the PDL as measured by dynamic mechanical analysis. Intriguingly, also noted was a weight gain as KAEKI mice aged. While several proteins associated with mineralized tissues are reported to affect energy metabolism, the metabolic role of the BSP-RGD region has yet to be elucidated. Here we focus on defining the role of the BSP-RGD region in metabolic activity. Methods Body weight, body composition, and caloric intake were measured in wild type (WT) and KAEKI mice. Energy expenditure was estimated using energy balance technique. Epididymal fat, interscapular fat, and liver were harvested for histological analysis. The systemic metabolic phenotype was assessed by sera analyses, insulin tolerance and glucose tolerance tests. Results The results showed that KAEKI mice developed mild obesity starting from 13 weeks postnatal (wpn). The increase in body weight correlated with an increase in lean mass and visceral adiposity. Histological examination revealed adipocyte hypertrophy in white epididymal fat and interscapular brown fat in KAEKI vs. WT mice at 17 wpn. Metabolic profiling indicated that KAEKI mice had dyslipidemia and hyperleptinemia but no significant changes in glucose metabolism. Energy balance analyses revealed that hyperphagia preceded weight gain in KAEKI mice. Conclusion These data suggest that the RGD region of BSP affects energy metabolism by regulating food intake, with further studies warranted to uncover the underlying mechanisms.
Collapse
Affiliation(s)
- Karin Nagasaki
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, United States
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Atsuhiro Nagasaki
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, United States
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Jocelyn M. Taylor
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Bernice 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, United States
| | - Yinyan Ma
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Martha 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, United States
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
5
|
Wölfel EM, von Kroge S, Matthies L, Koehne T, Petz K, Beikler T, Schmid-Herrmann CU, Kahl-Nieke B, Tsiakas K, Santer R, Muschol NM, Herrmann J, Busse B, Amling M, Rolvien T, Jandl NM, Barvencik F. Effects of Infantile Hypophosphatasia on Human Dental Tissue. Calcif Tissue Int 2023; 112:308-319. [PMID: 36414794 PMCID: PMC9968273 DOI: 10.1007/s00223-022-01041-4] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/02/2022] [Indexed: 11/23/2022]
Abstract
Hypophosphatasia (HPP) is an inherited, systemic disorder, caused by loss-of-function variants of the ALPL gene encoding the enzyme tissue non-specific alkaline phosphatase (TNSALP). HPP is characterized by low serum TNSALP concentrations associated with defective bone mineralization and increased fracture risk. Dental manifestations have been reported as the exclusive feature (odontohypophosphatasia) and in combination with skeletal complications. Enzyme replacement therapy (asfotase alfa) has been shown to improve respiratory insufficiency and skeletal complications in HPP patients, while its effects on dental status have been understudied to date. In this study, quantitative backscattered electron imaging (qBEI) and histological analysis were performed on teeth from two patients with infantile HPP before and during asfotase alfa treatment and compared to matched healthy control teeth. qBEI and histological methods revealed varying mineralization patterns in cementum and dentin with lower mineralization in HPP. Furthermore, a significantly higher repair cementum thickness was observed in HPP compared to control teeth. Comparison before and during treatment showed minor improvements in mineralization and histological parameters in the patient when normalized to matched control teeth. HPP induces heterogeneous effects on mineralization and morphology of the dental status. Short treatment with asfotase alfa slightly affects mineralization in cementum and dentin. Despite HPP being a rare disease, its mild form occurs at higher prevalence. This study is of high clinical relevance as it expands our knowledge of HPP and dental involvement. Furthermore, it contributes to the understanding of dental tissue treatment, which has hardly been studied so far.
Collapse
Affiliation(s)
- Eva Maria Wölfel
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
- Interdisciplinary Competence Center for Interface Research (ICCIR), Lottestr. 55A, Hamburg, Germany
| | - Simon von Kroge
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
| | - Levi Matthies
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Till Koehne
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
- Department of Orthodontics, Center for Dental and Oral Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Karin Petz
- Department of Periodontics, Preventive and Restorative Dentistry, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Thomas Beikler
- Department of Periodontics, Preventive and Restorative Dentistry, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Carmen Ulrike Schmid-Herrmann
- Department of Orthodontics, Center for Dental and Oral Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Bärbel Kahl-Nieke
- Department of Orthodontics, Center for Dental and Oral Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Konstantinos Tsiakas
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - René Santer
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Nicole Maria Muschol
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Jochen Herrmann
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, Section of Pediatric Radiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
- Interdisciplinary Competence Center for Interface Research (ICCIR), Lottestr. 55A, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
| | - Tim Rolvien
- Division of Orthopaedics, Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Nico Maximilian Jandl
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany
- Division of Orthopaedics, Department of Trauma and Orthopaedic Surgery, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Florian Barvencik
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 59, 22529, Hamburg, Germany.
| |
Collapse
|
6
|
Koh AJ, Nam HK, Michalski MN, Do J, McCauley LK, Hatch NE. Anabolic actions of parathyroid hormone in a hypophosphatasia mouse model. Osteoporos Int 2022; 33:2423-2433. [PMID: 35871207 PMCID: PMC9568459 DOI: 10.1007/s00198-022-06496-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 07/07/2022] [Indexed: 11/17/2022]
Abstract
UNLABELLED Hypophosphatasia, the rare heritable disorder caused by TNAP enzyme mutations, presents wide-ranging severity of bone hypomineralization and skeletal abnormalities. Intermittent PTH (1-34) increased long bone volume in Alpl-/- mice but did not alter the skull phenotype. PTH may have therapeutic value for adults with TNAP deficiency-associated osteoporosis. INTRODUCTION Hypophosphatasia is the rare heritable disorder caused by mutations in the tissue non-specific alkaline phosphatase (TNAP) enzyme leading to TNAP deficiency. Individuals with hypophosphatasia commonly present with bone hypomineralization and skeletal abnormalities. The purpose of this study was to determine the impact of intermittent PTH on the skeletal phenotype of TNAP-deficient Alpl-/- mice. METHODS Alpl-/- and Alpl+/+ (wild-type; WT) littermate mice were administered PTH (1-34) (50 µg/kg) or vehicle control from days 4 to 12 and skeletal analyses were performed including gross measurements, micro-CT, histomorphometry, and serum biochemistry. RESULTS Alpl-/- mice were smaller with shorter tibial length and skull length compared to WT mice. Tibial BV/TV was reduced in Alpl-/- mice and daily PTH (1-34) injections significantly increased BV/TV and BMD but not TMD in both WT and Alpl-/- tibiae. Trabecular spacing was not different between genotypes and was decreased by PTH in both genotypes. Serum P1NP was unchanged while TRAcP5b was significantly lower in Alpl-/- vs. WT mice, with no PTH effect, and no differences in osteoclast numbers. Skull height and width were increased in Alpl-/- vs. WT mice, and PTH increased skull width in WT but not Alpl-/- mice. Frontal skull bones in Alpl-/- mice had decreased BV/TV, BMD, and calvarial thickness vs. WT with no significant PTH effects. Lengths of cranial base bones (basioccipital, basisphenoid, presphenoid) and lengths of synchondroses (growth plates) between the cranial base bones, plus bone of the basioccipitus, were assessed. All parameters were reduced (except lengths of synchondroses, which were increased) in Alpl-/- vs. WT mice with no PTH effect. CONCLUSION PTH increased long bone volume in the Alpl-/- mice but did not alter the skull phenotype. These data suggest that PTH can have long bone anabolic activity in the absence of TNAP, and that PTH may have therapeutic value for individuals with hypophosphatasia-associated osteoporosis.
Collapse
Affiliation(s)
- Amy J Koh
- Department of Periodontology and Oral Medicine, 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
| | - Megan N Michalski
- Department of Cell Biology, Van Andel Institute, Grand Rapids, MI, USA
| | - Justin Do
- Department of Periodontology and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Laurie K McCauley
- Department of Periodontology and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
- Department of Pathology, School of Medicine, 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
|
7
|
Reibring CG, El Shahawy M, Hallberg K, Harfe BD, Linde A, Gritli-Linde A. Loss of BMP2 and BMP4 Signaling in the Dental Epithelium Causes Defective Enamel Maturation and Aberrant Development of Ameloblasts. Int J Mol Sci 2022; 23:6095. [PMID: 35682776 PMCID: PMC9180982 DOI: 10.3390/ijms23116095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/18/2022] [Accepted: 05/25/2022] [Indexed: 12/10/2022] Open
Abstract
BMP signaling is crucial for differentiation of secretory ameloblasts, the cells that secrete enamel matrix. However, whether BMP signaling is required for differentiation of maturation-stage ameloblasts (MA), which are instrumental for enamel maturation into hard tissue, is hitherto unknown. To address this, we used an in vivo genetic approach which revealed that combined deactivation of the Bmp2 and Bmp4 genes in the murine dental epithelium causes development of dysmorphic and dysfunctional MA. These fail to exhibit a ruffled apical plasma membrane and to reabsorb enamel matrix proteins, leading to enamel defects mimicking hypomaturation amelogenesis imperfecta. Furthermore, subsets of mutant MA underwent pathological single or collective cell migration away from the ameloblast layer, forming cysts and/or exuberant tumor-like and gland-like structures. Massive apoptosis in the adjacent stratum intermedium and the abnormal cell-cell contacts and cell-matrix adhesion of MA may contribute to this aberrant behavior. The mutant MA also exhibited severely diminished tissue non-specific alkaline phosphatase activity, revealing that this enzyme's activity in MA crucially depends on BMP2 and BMP4 inputs. Our findings show that combined BMP2 and BMP4 signaling is crucial for survival of the stratum intermedium and for proper development and function of MA to ensure normal enamel maturation.
Collapse
Affiliation(s)
- Claes-Göran Reibring
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, SE-40530 Göteborg, Sweden; (C.-G.R.); (M.E.S.); (K.H.); (A.L.)
| | - Maha El Shahawy
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, SE-40530 Göteborg, Sweden; (C.-G.R.); (M.E.S.); (K.H.); (A.L.)
- Department of Oral Biology, Faculty of Dentistry, Minia University, Minia 61511, Egypt
| | - Kristina Hallberg
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, SE-40530 Göteborg, Sweden; (C.-G.R.); (M.E.S.); (K.H.); (A.L.)
| | - Brian D. Harfe
- Department of Molecular Genetics and Microbiology Genetics Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Anders Linde
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, SE-40530 Göteborg, Sweden; (C.-G.R.); (M.E.S.); (K.H.); (A.L.)
| | - Amel Gritli-Linde
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, SE-40530 Göteborg, Sweden; (C.-G.R.); (M.E.S.); (K.H.); (A.L.)
| |
Collapse
|
8
|
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
|
9
|
Schroth RJ, Long C, Lee VHK, Alai-Towfigh H, Rockman-Greenberg C. Dental outcomes for children receiving asfotase alfa for hypophosphatasia. Bone 2021; 152:116089. [PMID: 34175501 DOI: 10.1016/j.bone.2021.116089] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/01/2021] [Accepted: 06/22/2021] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Hypophosphatasia, a genetic disease impeding development of teeth and bones, is associated with premature exfoliation of primary teeth. Hypophosphatasia is caused by mutations in the ALPL gene, which encodes the tissue non-specific form of alkaline phosphatase. Asfotase alfa (Strensiq®) is a human recombinant bone-targeted alkaline phosphatase. OBJECTIVES To review development and exfoliation patterns of primary/permanent teeth in a cohort of patients with hypophosphatasia enrolled in an open-label clinical trial of enzyme replacement therapy (ERT) with asfotase alfa. METHODS Data were collected from existing study files of a cohort of patients ≤5 years of age with infantile hypophosphatasia. Children were recruited at the Winnipeg site of a global clinical trial and were treated with ERT. Dental information, including the exfoliation/eruption patterns, were recorded at each visit. RESULTS Eleven children (7 females, 4 males) participated. Participants enrolled as infants (5 infants; mean age 3.0 ± 2.3 months) prematurely lost significantly fewer teeth to hypophosphatasia than patients recruited as preschoolers (6 preschoolers; mean age 52.5 ± 11.3 months), who started on asfotase alfa at a later age. Conclusion The oral health of children with early onset infantile hypophosphatasia may be improved with early and continued administration of ERT, compared to institution of therapy later in childhood.
Collapse
Affiliation(s)
- Robert J Schroth
- Rady Faculty of Health Sciences, University of Manitoba, Canada; Children's Hospital Research Institute of Manitoba, Canada; Section of Pediatric Dentistry, Winnipeg Regional Health Authority, Canada; Shared Health Manitoba, Canada.
| | - Catherine Long
- Rady Faculty of Health Sciences, University of Manitoba, Canada
| | - Victor H K Lee
- Rady Faculty of Health Sciences, University of Manitoba, Canada; Children's Hospital Research Institute of Manitoba, Canada
| | | | - Cheryl Rockman-Greenberg
- Rady Faculty of Health Sciences, University of Manitoba, Canada; Children's Hospital Research Institute of Manitoba, Canada; Shared Health Manitoba, Canada
| |
Collapse
|
10
|
Kinoshita Y, Mohamed FF, Amadeu de Oliveira F, Narisawa S, Miyake K, Foster BL, Millán JL. Gene Therapy Using Adeno-Associated Virus Serotype 8 Encoding TNAP-D 10 Improves the Skeletal and Dentoalveolar Phenotypes in Alpl -/- Mice. J Bone Miner Res 2021; 36:1835-1849. [PMID: 34076297 PMCID: PMC8446309 DOI: 10.1002/jbmr.4382] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 12/26/2022]
Abstract
Hypophosphatasia (HPP) is caused by loss-of-function mutations in the ALPL gene that encodes tissue-nonspecific alkaline phosphatase (TNAP), whose deficiency results in the accumulation of extracellular inorganic pyrophosphate (PPi ), a potent mineralization inhibitor. Skeletal and dental hypomineralization characterizes HPP, with disease severity varying from life-threatening perinatal or infantile forms to milder forms that manifest in adulthood or only affect the dentition. Enzyme replacement therapy (ERT) using mineral-targeted recombinant TNAP (Strensiq/asfotase alfa) markedly improves the life span, skeletal phenotype, motor function, and quality of life of patients with HPP, though limitations of ERT include frequent injections due to a short elimination half-life of 2.28 days and injection site reactions. We tested the efficacy of a single intramuscular administration of adeno-associated virus 8 (AAV8) encoding TNAP-D10 to increase the life span and improve the skeletal and dentoalveolar phenotypes in TNAP knockout (Alpl-/- ) mice, a murine model for severe infantile HPP. Alpl-/- mice received 3 × 1011 vector genomes/body of AAV8-TNAP-D10 within 5 days postnatal (dpn). AAV8-TNAP-D10 elevated serum ALP activity and suppressed plasma PPi . Treatment extended life span of Alpl-/- mice, and no ectopic calcifications were observed in the kidneys, aorta, coronary arteries, or brain in the 70 dpn observational window. Treated Alpl-/- mice did not show signs of rickets, including bowing of long bones, enlargement of epiphyses, or fractures. Bone microstructure of treated Alpl-/- mice was similar to wild type, with a few persistent small cortical and trabecular defects. Histology showed no measurable osteoid accumulation but reduced bone volume fraction in treated Alpl-/- mice versus controls. Treated Alpl-/- mice featured normal molar and incisor dentoalveolar tissues, with the exceptions of slightly reduced molar enamel and alveolar bone density. Histology showed the presence of cementum and normal periodontal ligament attachment. These results support gene therapy as a promising alternative to ERT for the treatment of HPP. © 2021 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)
- Yuka Kinoshita
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - 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
| | - Sonoko Narisawa
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Koichi Miyake
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Brian L Foster
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - José Luis Millán
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
11
|
Hasegawa A, Nakamura-Takahashi A, Kasahara M, Saso N, Narisawa S, Millán JL, Samura O, Sago H, Okamoto A, Umezawa A. Prenatal enzyme replacement therapy for Akp2 -/- mice with lethal hypophosphatasia. Regen Ther 2021; 18:168-175. [PMID: 34277899 PMCID: PMC8267436 DOI: 10.1016/j.reth.2021.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/28/2021] [Accepted: 06/06/2021] [Indexed: 11/21/2022] Open
Abstract
Hypophosphatasia (HPP) is a congenital skeletal disease. Impairment of bone mineralization and seizures are due to a deficiency of tissue-nonspecific alkaline phosphatase (TNAP). Enzyme replacement therapy (ERT) is available as a highly successful treatment for pediatric-onset HPP. However, the potential for prenatal ERT has not been fully investigated to date. In this study, we assessed outcomes and maternal safety using a combinational approach with prenatal and postnatal administration of recombinant TNAP in Akp2−/− mice as a model of infantile HPP. For the prenatal ERT, we administered subcutaneous injections of recombinant TNAP to pregnant mice from embryonic day 11.5–14.5 until delivery, and then sequentially to Akp2−/− pups from birth to day 18. For the postnatal ERT, we injected Akp2−/− pups from birth until day 18. Prenatal ERT did not cause any ectopic mineralization in heterozygous maternal mice. Both prenatal and postnatal ERT preserved growth, survival rate and improved bone calcification in Akp2−/− mice. However, the effects of additional prenatal treatment to newborn mice appeared to be minimal, and the difference between prenatal and postnatal ERT was subtle. Further improvement of the prenatal ERT schedule and long-term observation will be required. The present paper sets a standard for such future studies.
Collapse
Affiliation(s)
- Akihiro Hasegawa
- Center for Regenerative Medicine, National Center for Child Health and Development Research Institute, Tokyo, Japan.,Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | | | | | - Nana Saso
- Center for Regenerative Medicine, National Center for Child Health and Development Research Institute, Tokyo, Japan
| | - Sonoko Narisawa
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - José Luis Millán
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Osamu Samura
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Haruhiko Sago
- Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Akihiro Umezawa
- Center for Regenerative Medicine, National Center for Child Health and Development Research Institute, Tokyo, Japan
| |
Collapse
|
12
|
Su N, Zhu M, Cheng X, Xu K, Kocijan R, Zhang H. Six ALPL gene variants in five children with hypophosphatasia. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:888. [PMID: 34164522 PMCID: PMC8184488 DOI: 10.21037/atm-21-2096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Background Hypophosphatasia (HPP) is a rare hereditary disorder characterized by defective bone and tooth mineralization caused by mutations in the alkaline phosphatase (ALPL) gene encoding tissue-nonspecific alkaline phosphatase (TNSALP). Here we performed clinical and molecular studies on 5 HPP children to investigate the pathogenic mechanisms of the ALPL gene variants. Methods Clinical and genetic analyses were performed on 5 HPP children, and the loci where ALPL variants were identified. Plasmids containing the relevant loci were constructed. The molecular and cellular mechanisms of the pathogenic ALPL variants were investigated by cellular immunofluorescence, enzyme activity assay, and protein expression assay. Results A total of 6 ALPL variants were identified in 5 HPP children: proband 1: c.346G>A (p.A116T); proband 2: c.346G>A (p.A116T)/deletions from c.1097 to c.1099 CCT (p.T366_S367deli) compound heterozygous variant; proband 3: insertion of G from c.1014 to c.1015 (p.H338fs)/c.1446C>A (p.H482Q) compound heterozygous variant; proband 4: c.920C>T (p.P307L); and proband 5: c.883A>G (p.M295V). Twenty-four hours after the HEK-293T was transfected with different variant plasmids, its alkaline phosphatase activity and enzyme protein content were reduced compared with the wild type, and there were differences among different variants. Except for 1014-G-1015+C1446A, the degree of reduction in enzyme activity was negatively correlated with the severity of clinical manifestations. Immunofluorescence revealed that the variants (especially c.883A>G and c.920C>T) caused a decrease in alkaline phosphatase expression in the cellular membrane. Conclusions In total, 3 novel variants were identified in these 5 HPP children, the discovery of which will enrich the human ALPL gene mutation database. Different variants in the ALPL gene can downregulate the activity of TNSALP enzyme (and thus affect its function) by affecting protein expression and translational modifications. The same variant may cause clinical manifestations of different severities in different individuals due to the presence of dominant negative effects, alterations in noncoding sequences, blind area of intron regulatory region sequencing, and variations in environmental and individual factors. The molecular mechanisms via which the ALPL gene exerts its expression effect in vivo are highly variable and warrant further investigation.
Collapse
Affiliation(s)
- Na Su
- Department of Endocrinology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Department of Child Endocrinology and Genetic Metabolism, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Min Zhu
- Department of Endocrinology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xinran Cheng
- Department of Child Endocrinology and Genetic Metabolism, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ke Xu
- Department of Child Endocrinology and Genetic Metabolism, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Roland Kocijan
- Medical Faculty of Bone Diseases, Sigmund Freud University, Vienna, Austria.,Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of OEGK and AUVA Trauma Center Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria
| | - Huijiao Zhang
- Department of Endocrinology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
13
|
GSK3β rephosphorylation rescues ALPL deficiency-induced impairment of odontoblastic differentiation of DPSCs. Stem Cell Res Ther 2021; 12:225. [PMID: 33823913 PMCID: PMC8022410 DOI: 10.1186/s13287-021-02235-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/21/2021] [Indexed: 11/23/2022] Open
Abstract
Background Premature exfoliation of the deciduous teeth is a common manifestation in childhood patients with hypophosphatasia (HPP), which is an autosomal inherited disease caused by ALPL mutations. Dysplasia of the cementum, dentin, and alveolar bone has been proposed to be the main reasons for the exfoliation of teeth, while the extraordinarily complex intracellular mechanisms remain elusive. Dental pulp stem cells (DPSCs) have been demonstrated to successfully regenerate functional pulp-dentin-like tissue. Dental pulp cells derived from HPP patients impaired mineralization; however, insight into the deeper mechanism is still unclear. Methods The effects of ALPL on odontoblastic differentiation of DPSCs from HPP patient were assessed by Alizarin Red staining, immunofluorescent staining, Western blot and RT-PCR, and micro-CT assays. Result Here, we found DPSCs from HPP patient exhibited low ALP activity and impaired odontoblastic differentiation. Meanwhile, we found that loss of function of ALPL reduced phosphorylation of GSK3β in DPSCs. While GSK3β rephosphorylation improved odontoblastic differentiation of HPP DPSCs with LiCl treatment. Finally, we demonstrated systemic LiCl injection ameliorated tooth-associated defects in ALPL+/− mice by enhanced phosphorylation of GSK3β in the teeth. Conclusions Our study indicates that ALPL regulates odontoblastic differentiation of DPSCs and provides useful information for understanding how ALPL deficiency led to tooth dysplasia and, ultimately, may inform efforts at improvement tooth defects in HPP patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02235-7.
Collapse
|
14
|
Kramer K, Chavez MB, Tran AT, Farah F, Tan MH, Kolli TN, Dos Santos EJL, Wimer HF, Millán JL, Suva LJ, Gaddy D, Foster BL. Dental defects in the primary dentition associated with hypophosphatasia from biallelic ALPL mutations. Bone 2021; 143:115732. [PMID: 33160095 PMCID: PMC7769999 DOI: 10.1016/j.bone.2020.115732] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022]
Abstract
ALPL encodes tissue-nonspecific alkaline phosphatase (TNAP), an enzyme expressed in bone, teeth, liver, and kidney. ALPL loss-of-function mutations cause hypophosphatasia (HPP), an inborn error-of-metabolism that produces skeletal and dental mineralization defects. Case reports describe widely varying dental phenotypes, making it unclear how HPP comparatively affects the three unique dental mineralized tissues: enamel, dentin, and cementum. We hypothesized that HPP affected all dental mineralized tissues and aimed to establish quantitative measurements of dental tissues in a subject with HPP. The female proband was diagnosed with HPP during childhood based on reduced alkaline phosphatase activity (ALP), mild rachitic skeletal effects, and premature primary tooth loss. The diagnosis was subsequently confirmed genetically by the presence of compound heterozygous ALPL mutations (exon 5: c.346G>A, p.A116T; exon 10: c.1077C>G, p.I359M). Dental defects in 8 prematurely exfoliated primary teeth were analyzed by high resolution micro-computed tomography (micro-CT) and histology. Similarities to the Alpl-/- mouse model of HPP were identified by additional analyses of murine dentoalveolar tissues. Primary teeth from the proband exhibited substantial remaining root structure compared to healthy control teeth. Enamel and dentin densities were not adversely affected in HPP vs. control teeth. However, analysis of discrete dentin regions revealed an approximate 10% reduction in the density of outer mantle dentin of HPP vs. control teeth. All 4 incisors and the molar lacked acellular cementum by micro-CT and histology, but surprisingly, 2 of 3 prematurely exfoliated canines exhibited apparently normal acellular cementum. Based on dentin findings in the proband's teeth, we examined dentoalveolar tissues in a mouse model of HPP, revealing that the delayed initiation of mineralization in the incisor mantle dentin was associated with a broader lack of circumpulpal dentin mineralization. This study describes a quantitative approach to measure effects of HPP on dental tissues. This approach has uncovered a previously unrecognized novel mantle dentin defect in HPP, as well as a surprising and variable cementum phenotype within the teeth from the same HPP subject.
Collapse
Affiliation(s)
- K Kramer
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - M B Chavez
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - A T Tran
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - F Farah
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - M H Tan
- 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
| | - E J Lira Dos Santos
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA; Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas-UNICAMP, Piracicaba, SP, Brazil
| | - H F Wimer
- Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA; National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - J L Millán
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - L J Suva
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - D Gaddy
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - B L Foster
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
15
|
Inhibitory Effect of Lactobacillus plantarum CCFM8724 towards Streptococcus mutans- and Candida albicans-Induced Caries in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2020:4345804. [PMID: 33414892 PMCID: PMC7769668 DOI: 10.1155/2020/4345804] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/26/2020] [Accepted: 12/06/2020] [Indexed: 11/17/2022]
Abstract
Streptococcus mutans is a recognized cariogenic bacterium and a major producer of biofilm matrix. The presence of Candida albicans in dental plaque with S. mutans enhances the virulence leading to the onset of rampant caries which is similar to early childhood caries (ECC). The purpose of this study was to explore the effect of Lactobacillus plantarum CCFM8724 (CCFM8724) on the treatment and prevention of dental caries induced by S. mutans and C. albicans in vivo. Rats were divided into 6 groups: the control group and model group, 2 treatment groups, and 2 prevention groups (0.02% chlorhexidine or CCFM8724). The fluctuation of microbial colonization and the change of bacteria flora in rat oral cavity after sowing of L. plantarum CCFM8724 were investigated by colony-forming units (CFU) and microflora analysis. The caries of rats were assessed by microcomputed tomography (micro-CT) and Keyes scoring method. The results showed that L. plantarum CCFM8724 in both the treatment and prevention groups could significantly decrease the population of S. mutans and C. albicans in the rats' oral cavity (p < 0.001), the mineral loss of enamel (p < 0.05), and the scores of caries (p < 0.05). Besides, L. plantarum CCFM8724 exhibited better effects than chlorhexidine. Hence, L. plantarum CCFM8724 was proved to be a potential oral probiotic on caries treatment and prevention in vivo and it may have the prospect of application in dental caries (especially ECC) prevention products.
Collapse
|
16
|
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
|
17
|
Yoshizaki K, Fukumoto S, Bikle DD, Oda Y. Transcriptional Regulation of Dental Epithelial Cell Fate. Int J Mol Sci 2020; 21:ijms21238952. [PMID: 33255698 PMCID: PMC7728066 DOI: 10.3390/ijms21238952] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 12/28/2022] Open
Abstract
Dental enamel is hardest tissue in the body and is produced by dental epithelial cells residing in the tooth. Their cell fates are tightly controlled by transcriptional programs that are facilitated by fate determining transcription factors and chromatin regulators. Understanding the transcriptional program controlling dental cell fate is critical for our efforts to build and repair teeth. In this review, we describe the current understanding of these regulators essential for regeneration of dental epithelial stem cells and progeny, which are identified through transgenic mouse models. We first describe the development and morphogenesis of mouse dental epithelium in which different subpopulations of epithelia such as ameloblasts contribute to enamel formation. Then, we describe the function of critical factors in stem cells or progeny to drive enamel lineages. We also show that gene mutations of these factors are associated with dental anomalies in craniofacial diseases in humans. We also describe the function of the master regulators to govern dental lineages, in which the genetic removal of each factor switches dental cell fate to that generating hair. The distinct and related mechanisms responsible for the lineage plasticity are discussed. This knowledge will lead us to develop a potential tool for bioengineering new teeth.
Collapse
Affiliation(s)
- Keigo Yoshizaki
- Section of Orthodontics and Dentofacial Orthopedics, Division of Oral Health, Growth and Development, Kyushu University Faculty of Dental Science, Fukuoka 812-8582, Japan;
| | - Satoshi Fukumoto
- Section of Pediatric Dentistry, Division of Oral Health, Growth and Development, Kyushu University Faculty of Dental Science, Fukuoka 812-8582, Japan;
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Daniel D. Bikle
- Departments of Medicine and Endocrinology, University of California San Francisco and Veterans Affairs Medical Center, San Francisco, CA 94158, USA;
| | - Yuko Oda
- Departments of Medicine and Endocrinology, University of California San Francisco and Veterans Affairs Medical Center, San Francisco, CA 94158, USA;
- Correspondence:
| |
Collapse
|
18
|
Machiya A, Tsukamoto S, Ohte S, Kuratani M, Suda N, Katagiri T. Smad4-dependent transforming growth factor-β family signaling regulates the differentiation of dental epithelial cells in adult mouse incisors. Bone 2020; 137:115456. [PMID: 32473314 DOI: 10.1016/j.bone.2020.115456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 01/22/2023]
Abstract
Teeth consist of two major tissues, enamel and dentin, which are formed during development by epithelial and mesenchymal cells, respectively. Rodent incisors are useful experimental models for studying the molecular mechanisms of tooth formation because they are simultaneously growing in not only embryos but also adults. Members of the transforming growth factor-β (TGF-β) family regulate epithelial-mesenchymal interactions through an essential coactivator, Smad4. In the present study, we established Smad4 conditional knockout (cKO) mice and examined phenotypes in adult incisors. Smad4 cKO mice died with severe anemia within one month. Phosphorylated Smad1/5/9 and Smad2/3 were detected in epithelial cells in both control and Smad4 cKO mice. Disorganized and hypoplastic epithelial cells, such as ameloblasts, were observed in Smad4 cKO mice. Moreover, alkaline phosphatase expression and iron accumulation were reduced in dental epithelial cells in Smad4 cKO mice. These findings suggest that TGF-β family signaling through Smad4 is required for the differentiation and functions of dental epithelial cells in adult mouse incisors.
Collapse
Affiliation(s)
- Aiko Machiya
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan; Division of Orthodontics, Department of Human Development and Fostering, Meikai University School of Dentistry, Saitama, Japan; Division of Oral Rehabilitation of Sciences, Department of Restorative and Biomaterials Sciences, Meikai University School of Dentistry, Saitama, Japan
| | - Sho Tsukamoto
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Satoshi Ohte
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan; Microbial Chemistry Laboratory, Graduate School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan
| | - Mai Kuratani
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Naoto Suda
- Division of Orthodontics, Department of Human Development and Fostering, Meikai University School of Dentistry, Saitama, Japan
| | - Takenobu Katagiri
- Division of Biomedical Sciences, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan.
| |
Collapse
|
19
|
Bao J, Song Z, Song C, Wang Y, Li W, Mai W, Shi Q, Yu H, Ni L, Liu Y, Lu X, He C, Chen L, Qu G. Identification of Biomarkers for Osteosarcoma Based on Integration Strategy. Med Sci Monit 2020; 26:e920803. [PMID: 32173717 PMCID: PMC7101204 DOI: 10.12659/msm.920803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Background Osteosarcoma (OS) is the most common primary malignant tumor of bone. The identification of novel biomarkers is necessary for the diagnosis and treatment of osteosarcoma. Material/Methods We obtained 11 paired fresh-frozen OS samples and normal controls from patients between September 2015 and February 2017. We used an integration strategy that analyzes next-generation sequencing data by bioinformatics methods based on the pathogenesis of osteosarcoma. Results One susceptibility lncRNA and 7 susceptibility genes regulated by the lncRNA for osteosarcoma were effectively identified, and real-time PCR and clinical index ALP data were used to test their effectiveness. Conclusions The results showed that the expression levels of the 7 genes were highly consistent in the training and test sample sets, especially between the expression value of the gene ALPL and the plasma detection value of its encoded protein ALP. In particular, both the expression of gene ALPL and the plasma detection values of protein ALP encoded by gene ALPL showed a high degree of consistency among different data types. The identified lncRNA and genes effectively classified the samples proved so that they could be used as potential biomarkers of osteosarcoma. Our strategy may also be helpful for the identification of biomarkers for other diseases.
Collapse
Affiliation(s)
- Junjie Bao
- Department of Orthopedic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China (mainland)
| | - Zhaona Song
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Chunyu Song
- Department of Orthopedic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China (mainland)
| | - Yahui Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Wan Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Wei Mai
- Department of Orthopedic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China (mainland)
| | - Qingyu Shi
- Department of Orthopedic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China (mainland)
| | - Hongwei Yu
- Department of Orthopedic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China (mainland)
| | - Linying Ni
- Department of Orthopedic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China (mainland)
| | - Yishu Liu
- Department of Orthopedic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China (mainland)
| | - Xiaolin Lu
- Department of Orthopedic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China (mainland)
| | - Chuan He
- Department of Orthopedic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China (mainland)
| | - Lina Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Guofan Qu
- Department of Orthopedic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China (mainland)
| |
Collapse
|
20
|
Di Rocco F, Rothenbuhler A, Cormier Daire V, Bacchetta J, Adamsbaum C, Baujat G, Rossi M, Lingart A. Craniosynostosis and metabolic bone disorder. A review. Neurochirurgie 2019; 65:258-263. [PMID: 31562881 DOI: 10.1016/j.neuchi.2019.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/12/2019] [Accepted: 09/15/2019] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Some metabolic bone disorders may result in the premature closure of one or more calvarial sutures during childhood, potentially leading to a cranioencephalic disproportion. The aim of this paper is to review the characteristics and consequences of craniosynostosis associated with metabolic disorder. MATERIAL AND METHODS A review of the literature on metabolic forms of craniosynostosis was performed. RESULTS The most common forms of craniosynostosis associated with metabolic bone disorder were isolated sagittal suture fusion with or without scaphocephaly, and sagittal suture fusion associated with coronal suture fusion (oxycephaly) or also with lambdoid suture fusion (pansynostosis). Synostosis may be well-tolerated, but in some subjects results in neurodevelopmental and functional impairment that is sometimes severe. CONCLUSION The impact of metabolic synostosis is very variable, depending on the specific underlying metabolic disease, with a large spectrum of morphological and functional consequences. Diagnosis should be early and management should be carried out by a multidisciplinary team with expertise in both rare skeletal disorders and craniosynostosis. The impact of emergent medical therapies recently developed for some of these diseases will be assessed by systematic coherent follow-up of international registries.
Collapse
Affiliation(s)
- F Di Rocco
- Inserm 1033, neurochirurgie pédiatrique, centre de référence pour les craniosténoses, Lyon et université Claude Bernard Lyon 1, hôpital femme-mère-enfant, 69003 Lyon, France.
| | - A Rothenbuhler
- Endocrinologie et diabète de l'enfant, filière OSCAR et plateforme d'expertise Paris Sud maladies rares, centre de référence des maladies rares du calcium et du phosphate, hôpital Bicêtre Paris Sud, AP-HP, 94270 Le Kremlin Bicêtre, France
| | - V Cormier Daire
- Centre de référence maladies osseuses constitutionnelles, institut imagine, 75015 Paris, France
| | - J Bacchetta
- Inserm 1033, centre de référence des maladies rares du calcium et du phosphate, université Claude Bernard Lyon 1, hôpital femme-mère-enfant, 69003 Lyon, France
| | - C Adamsbaum
- Service de radiologie pédiatrique, université Paris-Saclay, hôpital Bicêtre, AP-HP, 94270 Le Kremlin Bicêtre, France
| | - G Baujat
- Centre de référence maladies osseuses constitutionnelles, institut imagine, 75015 Paris, France
| | - M Rossi
- GENDEV Team, CNRS UMR5292, CRNL, UCBL1, Inserm U1028, service de génétique, centre de référence anomalies du développement, centre de compétence maladies osseuses constitutionnelles, hospices civils de Lyon, 69003 Lyon, France
| | - A Lingart
- Endocrinologie et diabète de l'enfant, filière OSCAR et plateforme d'expertise Paris Sud maladies rares, centre de référence des maladies rares du calcium et du phosphate, hôpital Bicêtre Paris Sud, AP-HP, 94270 Le Kremlin Bicêtre, France; Service de radiologie pédiatrique, université Paris-Saclay, hôpital Bicêtre, AP-HP, 94270 Le Kremlin Bicêtre, France; Inserm U1185, université Paris Sud Paris-Saclay, 94270 Le Kremlin Bicêtre, France
| |
Collapse
|
21
|
Hypophosphatasia and the importance of the general dental practitioner - a case series and discussion of upcoming treatments. Br Dent J 2019; 224:937-943. [PMID: 29999027 DOI: 10.1038/sj.bdj.2018.441] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2018] [Indexed: 01/07/2023]
Abstract
Hypophosphatasia (HPP) is an inherited metabolic disorder that results in poorly mineralised bones and teeth. Clinical symptoms vary widely from mild dental anomalies to fatal fetal defects. The most common dental symptoms include exfoliation of the primary incisors before the age of three with little or no root resorption, large pulp chambers, alveolar bone loss and thin dentinal walls. There is generally minimal periodontal inflammation associated with the bony destruction and tooth loss. The general dental practitioner is usually the first clinician to spot signs of the milder forms of HPP. Patients diagnosed with dental symptoms in childhood can go on to develop significant morbidity in middle age with chronic bone pain and stress fractures of the long bones. The primary dental care clinician is the key to early diagnosis of such cases, whether they present in childhood or adulthood. Emerging enzyme replacement therapy has considerably changed the landscape of the disease, resulting in astonishing improvements in bone mineralisation and a significant reduction in mortality and morbidity. It is increasingly likely that primary and secondary care clinicians will treat patients with the severe forms of HPP, who would previously not have survived infancy.
Collapse
|
22
|
Yin L, Chang Y, You Y, Liu C, Li J, Lai HC. Biological responses of human bone mesenchymal stem cells to Ti and TiZr implant materials. Clin Implant Dent Relat Res 2019; 21:550-564. [PMID: 31009155 DOI: 10.1111/cid.12756] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 01/06/2019] [Accepted: 01/29/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Titanium-zirconium alloy (TiZr1317) is a new material used for biological implants. There are several studies on the effects of TiZr implants on the biological characteristics of human bone mesenchymal stem cells (hBMSCs). PURPOSE The purpose of this study was to investigate the biological responses of hBMSCs to implant holes affected by the physicochemical properties of oral implants (TiSLA , TiSLActive , TiZrSLA , and TiZrSLActive ). MATERIALS AND METHODS Grade 4 Ti and TiZr (13-17% Zr) substrates were modified by sand-blasted large-grit acid-etched (SLA) or hydrophilic sand-blasted large-grit acid-etched (SLActive), resulting in four types of surface with complex microstructures corresponding to the commercially-available implants SLA, RoxolidSLA, SLActive, and RoxolidSLActive (Institute Straumann AG, Basel, Switzerland). Physicochemical properties were detected and the biological responses of hBMSCs were observed. RESULTS Surface morphology characterization by scanning electron microscopy and atomic force microscopy revealed differences between the four groups. SLActive had higher surface energy/wettability than SLA, indicating that increased surface energy/wettability can promote the absorption of osteogenic proteins and enhance osseointegration. hBMSCs seeded on SLActive substrates exhibited better performance in terms of cell attachment, proliferation and osteoblastic differentiation than cells seeded on SLA. CONCLUSION Because of their more suitable physicochemical properties, TiSLActive and TiZrSLActive materials demonstrated more pronounced effects on the biological responses of hBMSCs compared with TiSLA and TiZrSLA .
Collapse
Affiliation(s)
- Lihua Yin
- Department of Implant Dentistry, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai JiaoTong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yaoren Chang
- Department of Implantology, School/Hospital of Stomatology Lanzhou University, Lanzhou, Gansu, China
| | - Yuanhe You
- Department of Implantology, School/Hospital of Stomatology Lanzhou University, Lanzhou, Gansu, China
| | - Chun Liu
- Department of Implantology, School/Hospital of Stomatology Lanzhou University, Lanzhou, Gansu, China
| | - Jie Li
- Department of Implantology, School/Hospital of Stomatology Lanzhou University, Lanzhou, Gansu, China
| | - Hong-Chang Lai
- Department of Implant Dentistry, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai JiaoTong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Abstract
PURPOSE OF REVIEW Hypophosphatasia (HPP) is a rare genetic disorder caused by mutations of the ALPL gene. ALPL encodes the tissue-non-specific isoenzyme of alkaline phosphatase (TNSALP). Consequently, bone mineralization is decreased leading to fractures, arthralgia, and extra-skeletal manifestations including tissue calcification, respiratory failure, and neurological complications. This review summarizes the most important clinical findings, diagnosis, and treatment options for HPP. RECENT FINDINGS Asfotase alfa is a recombinant human alkaline phosphatase, used as treatment for the underlying cause of HPP. Asfotase alfa enhances the survival in life-threatening HPP and improves bone mineralization, muscle strength, and pulmonary function. However, discontinuation of asfotase alfa leads to reappearance of bone hypomineralization. Due to its varied manifestations, HPP often mimics rheumatological and other bone diseases, thereby delaying its diagnosis. Asfotase alfa, a recombinant alkaline phosphatase, is available for the long-term enzyme replacement therapy in patients with pediatric-onset HPP to treat the bone manifestations of the disease.
Collapse
|
26
|
Bover J, Ureña P, Aguilar A, Mazzaferro S, Benito S, López-Báez V, Ramos A, daSilva I, Cozzolino M. Alkaline Phosphatases in the Complex Chronic Kidney Disease-Mineral and Bone Disorders. Calcif Tissue Int 2018; 103:111-124. [PMID: 29445837 DOI: 10.1007/s00223-018-0399-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/29/2018] [Indexed: 12/16/2022]
Abstract
Alkaline phosphatases (APs) remove the phosphate (dephosphorylation) needed in multiple metabolic processes (from many molecules such as proteins, nucleotides, or pyrophosphate). Therefore, APs are important for bone mineralization but paradoxically they can also be deleterious for other processes, such as vascular calcification and the increasingly known cross-talk between bone and vessels. A proper balance between beneficial and harmful activities is further complicated in the context of chronic kidney disease (CKD). In this narrative review, we will briefly update the complexity of the enzyme, including its different isoforms such as the bone-specific alkaline phosphatase or the most recently discovered B1x. We will also analyze the correlations and potential discrepancies with parathyroid hormone and bone turnover and, most importantly, the valuable recent associations of AP's with cardiovascular disease and/or vascular calcification, and survival. Finally, a basic knowledge of the synthetic and degradation pathways of APs promises to open new therapeutic strategies for the treatment of the CKD-Mineral and Bone Disorder (CKD-MBD) in the near future, as well as for other processes such as sepsis, acute kidney injury, inflammation, endothelial dysfunction, metabolic syndrome or, in diabetes, cardiovascular complications. However, no studies have been done using APs as a primary therapeutic target for clinical outcomes, and therefore, AP's levels cannot yet be used alone as an isolated primary target in the treatment of CKD-MBD. Nonetheless, its diagnostic and prognostic potential should be underlined.
Collapse
Affiliation(s)
- Jordi Bover
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, RedinRen, C. Cartagena, Catalonia, 340-350, Barcelona, Spain.
| | - Pablo Ureña
- Department of Nephrology and Dialysis, Clinique du Landy and Department of Renal Physiology, Necker Hospital, University of Paris Descartes, Paris, France
| | - Armando Aguilar
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, RedinRen, C. Cartagena, Catalonia, 340-350, Barcelona, Spain
| | - Sandro Mazzaferro
- Department of Cardiovascular, Respiratory, Nephrologic and Geriatric Sciences, Sapienza University of Rome, Rome, Italy
| | - Silvia Benito
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, RedinRen, C. Cartagena, Catalonia, 340-350, Barcelona, Spain
| | - Víctor López-Báez
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, RedinRen, C. Cartagena, Catalonia, 340-350, Barcelona, Spain
| | - Alejandra Ramos
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, RedinRen, C. Cartagena, Catalonia, 340-350, Barcelona, Spain
| | - Iara daSilva
- Department of Nephrology, Fundació Puigvert, IIB Sant Pau, RedinRen, C. Cartagena, Catalonia, 340-350, Barcelona, Spain
| | - Mario Cozzolino
- Laboratory of Experimental Nephrology, Renal Division,San Paolo Hospital, DiSS University of Milan, Milan, Italy
| |
Collapse
|
27
|
Greene SL, Mamaeva O, Crossman DK, Lu C, MacDougall M. Gene-Expression Analysis Identifies IGFBP2 Dysregulation in Dental Pulp Cells From Human Cleidocranial Dysplasia. Front Genet 2018; 9:178. [PMID: 29875795 PMCID: PMC5974155 DOI: 10.3389/fgene.2018.00178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 04/30/2018] [Indexed: 12/04/2022] Open
Abstract
Cleidocranial dysplasia (CCD) is an autosomal dominant disorder affecting osteoblast differentiation, chondrocyte maturation, skeletal morphogenesis, and tooth formation. Dental phenotype in CCD include over-retained primary teeth, failed eruption of permanent teeth, and supernumerary teeth. The underlying mechanism is unclear. We previously reported one CCD patient with allelic RUNX2 deletion (CCD-011). In the current study, we determined the transcriptomic profiles of dental pulp cells from this patient compared to one sex-and-age matched non-affected individual. Next Generation RNA sequencing revealed that 60 genes were significantly dysregulated (63% upregulated and 27% downregulated). Among them, IGFBP2 (insulin-like growth factor binding protein-2) was found to be upregulated more than twofold in comparison to control cells. Stable overexpression of RUNX2 in CCD-011 pulp cells resulted in the reduction of IGFBP2. Moreover, ALPL expression was up-regulated in CCD-011 pulp cells after introduction of normal RUNX2. Promoter analysis revealed that there are four proximal putative RUNX2 binding sites in -1.5 kb IGFBP2 promoter region. Relative luciferase assay confirmed that IGFBP2 is a direct target of RUNX2. Immunohistochemistry demonstrated that IGFBP2 was expressed in odontoblasts but not ameloblasts. This report demonstrated the importance of RUNX2 in the regulation of gene profile related to dental pulp cells and provided novel insight of RUNX2 into the negative regulation of IGFBP2.
Collapse
Affiliation(s)
- Stephen L. Greene
- Department of Pediatric Dentistry, School of Dentistry, The University of Alabama at Birmingham, Birmingham, AL, United States
- Institute of Oral Health Research, School of Dentistry, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Olga Mamaeva
- Institute of Oral Health Research, School of Dentistry, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - David K. Crossman
- Department of Genetics, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Changming Lu
- Institute of Oral Health Research, School of Dentistry, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mary MacDougall
- Faculty of Dentistry, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
28
|
Abstract
We review here clinical, pathophysiological, diagnostic, genetic and molecular aspects of Hypophosphatasia (HPP), a rare inherited metabolic disorder. The clinical presentation is a continuum ranging from a prenatal lethal form with no skeletal mineralization to a mild form with late adult onset presenting with nonpathognomonic symptoms. The prevalence of severe forms is low, whereas less severe forms are more frequently observed. The disease is caused by loss-of-function mutations in the ALPL gene encoding the Tissue Nonspecific Alkaline Phosphatase (TNSALP), a central regulator of mineralization. Severe forms are recessively inherited, whereas moderate forms are either recessively or dominantly inherited, and the more severe the disease is, the more often it is subject to recessive inheritance. The diagnosis is based on a constantly low alkaline phosphatase (AP) activity in serum and genetic testing that identifies ALPL mutations. More than 340 mutations have been identified and are responsible for the extraordinary clinical heterogeneity. A clear but imperfect genotype-phenotype correlation has been observed, suggesting that other genetic or environmental factors modulate the phenotype. Enzyme replacement therapy is now available for HPP, and other approaches, such as gene therapy, are currently being investigated.
Collapse
Affiliation(s)
- Etienne Mornet
- Unité de Génétique Constitutionnelle, Service de Biologie, Centre Hospitalier de Versailles, 177 rue de Versailles, 78150 Le Chesnay, France.
| |
Collapse
|
29
|
Zhang TT, Guo HJ, Liu XJ, Chu JP, Zhou XD. Galla chinensis Compounds Remineralize Enamel Caries Lesions in a Rat Model. Caries Res 2017; 50:159-65. [PMID: 27111668 DOI: 10.1159/000445036] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 02/26/2016] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES To investigate the effect of Galla chinensis chemical compounds on enamel caries remineralization in rats. METHODS A total of 40 rats were inoculated with Streptococcus sobrinus 6715 and fed a cariogenic diet (Keyes 2000). The rats were randomly divided into 4 groups and treated topically twice a day with each of the following extracts (or control) for 5 weeks: distilled and deionized water (DDW, negative control); 1,000 ppm NaF (positive control); 4,000 ppm G. chinensis crude aqueous extract (GCE), or 4,000 ppm gallic acid (GA). After the experimental period, Keyes' caries diagnosis and scoring technique was applied as a preliminary evaluation on the molar teeth. For more accurate remineralization data, the residual enamel volume of the first molar and the mineral density (MD) of the enamel were further analyzed by micro-CT. RESULTS The severity of molar enamel caries decreased in the following order of treatment groups: DDW > GA > GCE > NaF (p < 0.05). In addition, the residual first molar enamel volume and MD values increased in the order of DDW, GA, GCE and NaF treatment groups (p < 0.05). In most molars, remineralization layers were observed on the surface of lesions. CONCLUSION G. chinensis compounds remineralize enamel caries lesions in a rat model.
Collapse
Affiliation(s)
- Tie-Ting Zhang
- College of Stomatology, Zhengzhou University, Zhengzhou, China
| | | | | | | | | |
Collapse
|
30
|
Abstract
Hypophosphatasia (HPP) is the inborn-error-of-metabolism that features low serum alkaline phosphatase (ALP) activity (hypophosphatasemia) caused by loss-of-function mutation(s) of the gene that encodes the tissue-nonspecific isoenzyme of ALP (TNSALP). Autosomal recessive or autosomal dominant inheritance from among >300 TNSALP (ALPL) mutations largely explains HPP's remarkably broad-ranging severity. TNSALP is a cell-surface homodimeric phosphohydrolase richly expressed in the skeleton, liver, kidney, and developing teeth. In HPP, TNSALP substrates accumulate extracellularly. Among them is inorganic pyrophosphate (PPi), a potent inhibitor of mineralization. Superabundance of extracellular PPi explains the hard tissue complications of HPP that feature premature loss of deciduous teeth and often rickets or osteomalacia as well as calcific arthropathies in some affected adults. In infants with severe HPP, blocked entry of minerals into the skeleton can cause hypercalcemia, and insufficient hydrolysis of pyridoxal 5'-phosphate (PLP), the major circulating form of vitamin B6, can cause pyridoxine-dependent seizures. Elevated circulating PLP is a sensitive and specific biochemical marker for HPP. Also, the TNSALP substrate phosphoethanolamine (PEA) is usually elevated in serum and urine in HPP, though less reliably for diagnosis. Pathognomonic radiographic changes occur in pediatric HPP when the skeletal disease is severe. TNSALP mutation analysis is essential for recurrence risk assessment for HPP in future pregnancies and for prenatal diagnosis. HPP was the final rickets/osteomalacia to have a medical treatment. Now, significant successes using asfotase alfa, a mineral-targeted recombinant TNSALP, are published concerning severely affected newborns, infants, and children. Asfotase alfa was approved by regulatory agencies multinationally in 2015 typically for pediatric-onset HPP.
Collapse
Affiliation(s)
- Michael P Whyte
- Center for Metabolic Bone Disease and Molecular Research, Shriners Hospital for Children, Division of Bone and Mineral Diseases, Department of Internal Medicine, Washington University School of Medicine at Barnes-Jewish Hospital, St. Louis, Missouri, USA.
| |
Collapse
|
31
|
Yoshizaki K, Hu L, Nguyen T, Sakai K, Ishikawa M, Takahashi I, Fukumoto S, DenBesten PK, Bikle DD, Oda Y, Yamada Y. Mediator 1 contributes to enamel mineralization as a coactivator for Notch1 signaling and stimulates transcription of the alkaline phosphatase gene. J Biol Chem 2017; 292:13531-13540. [PMID: 28673966 DOI: 10.1074/jbc.m117.780866] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/17/2017] [Indexed: 12/21/2022] Open
Abstract
Tooth enamel is mineralized through the differentiation of multiple dental epithelia including ameloblasts and the stratum intermedium (SI), and this differentiation is controlled by several signaling pathways. Previously, we demonstrated that the transcriptional coactivator Mediator 1 (MED1) plays a critical role in enamel formation. For instance, conditional ablation of Med1 in dental epithelia causes functional changes in incisor-specific dental epithelial stem cells, resulting in mineralization defects in the adult incisors. However, the molecular mechanism by which Med1 deficiency causes these abnormalities is not clear. Here, we demonstrated that Med1 ablation causes early SI differentiation defects resulting in enamel hypoplasia of the Med1-deficient molars. Med1 deletion prevented Notch1-mediated differentiation of the SI cells resulting in decreased alkaline phosphatase (ALPL), which is essential for mineralization. However, it does not affect the ability of ameloblasts to produce enamel matrix proteins. Using the dental epithelial SF2 cell line, we demonstrated that MED1 directly activates transcription of the Alpl gene through the stimulation of Notch1 signaling by forming a complex with cleaved Notch1-RBP-Jk on the Alpl promoter. These results suggest that MED1 may be essential for enamel matrix mineralization by serving as a coactivator for Notch1 signaling regulating transcription of the Alpl gene.
Collapse
Affiliation(s)
- Keigo Yoshizaki
- From the Laboratory of Cell and Developmental Biology, NIDCR, National Institutes of Health, Bethesda, Maryland 20892.,the Division of Oral Health, Growth, and Development, Kyushu University Faculty of Dental Science, Fukuoka 812-8582, Japan
| | - Lizhi Hu
- the Departments of Medicine and Dermatology, the University of California San Francisco and the Veterans Affairs Medical Center, San Francisco, California 94158
| | - Thai Nguyen
- the Departments of Medicine and Dermatology, the University of California San Francisco and the Veterans Affairs Medical Center, San Francisco, California 94158
| | - Kiyoshi Sakai
- From the Laboratory of Cell and Developmental Biology, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Masaki Ishikawa
- From the Laboratory of Cell and Developmental Biology, NIDCR, National Institutes of Health, Bethesda, Maryland 20892
| | - Ichiro Takahashi
- the Division of Oral Health, Growth, and Development, Kyushu University Faculty of Dental Science, Fukuoka 812-8582, Japan
| | - Satoshi Fukumoto
- the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan, and
| | - Pamela K DenBesten
- the University of California San Francisco School of Dentistry, San Francisco, California 94143
| | - Daniel D Bikle
- the Departments of Medicine and Dermatology, the University of California San Francisco and the Veterans Affairs Medical Center, San Francisco, California 94158
| | - Yuko Oda
- the Departments of Medicine and Dermatology, the University of California San Francisco and the Veterans Affairs Medical Center, San Francisco, California 94158,
| | - Yoshihiko Yamada
- From the Laboratory of Cell and Developmental Biology, NIDCR, National Institutes of Health, Bethesda, Maryland 20892,
| |
Collapse
|
32
|
Di Rocco F, Baujat G, Cormier-Daire V, Rothenbuhler A, Linglart A. Craniosynostosis and hypophosphatasia. Arch Pediatr 2017; 24:5S89-5S92. [DOI: 10.1016/s0929-693x(18)30022-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
33
|
Abstract
BACKGROUND Hypophosphatasia (HPP) is a rare inherited metabolic disease in which mutations in the ALPL gene (encoding tissue-nonspecific alkaline phosphatase) result in varying degrees of enzyme deficiency. HPP manifests in a spectrum of symptoms, including early primary tooth loss (root intact) and alveolar bone mineralisation defects. OBJECTIVE To provide an overview of HPP for dental professionals to help recognise and differentially diagnose patients for appropriate referral to a specialist team. METHODS A non-systematic review of publications on HPP was performed. RESULTS Different forms of HPP are described, along with characteristic symptoms and laboratory findings. Diagnosis is challenging due to the rareness and variable presentation of symptoms. Low alkaline phosphatase levels are a signature of HPP, but reference ranges vary according to gender and age. Key features are defined and management strategies discussed, focusing on enzyme replacement therapy. Finally, a patient registry aimed at better defining the prevalence of HPP and raising awareness is described. CONCLUSIONS HPP is a rare disease with a wide spectrum of manifestations, with orodental symptoms featuring prominently in the natural history. Dental professionals may be positioned at the beginning of the diagnostic pathway; thus, recognition of HPP features for timely referral and optimal disease management is important.
Collapse
Affiliation(s)
- Agnès Bloch-Zupan
- Faculté de Chirurgie Dentaire, Université de Strasbourg, Strasbourg, France.
- Hôpitaux Universitaires de Strasbourg, Pôle de Médecine et Chirurgie Bucco-Dentaires, Centre de référence des manifestations odontologiques des maladies rares (CRMR), Reference Centre for Orodental Manifestations of Rare Diseases, Strasbourg, France.
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS-UdS UMR7104, Université de Strasbourg, Illkirch, France.
- Eastman Dental Institute, University College London, London, UK.
| |
Collapse
|
34
|
Foster BL, Kuss P, Yadav MC, Kolli TN, Narisawa S, Lukashova L, Cory E, Sah RL, Somerman MJ, Millán JL. Conditional Alpl Ablation Phenocopies Dental Defects of Hypophosphatasia. J Dent Res 2016; 96:81-91. [PMID: 27582029 DOI: 10.1177/0022034516663633] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Loss-of-function mutations in ALPL result in hypophosphatasia (HPP), an inborn error of metabolism that causes defective skeletal and dental mineralization. ALPL encodes tissue-nonspecific alkaline phosphatase, an enzyme expressed in bone, teeth, liver, and kidney that hydrolyzes the mineralization inhibitor inorganic pyrophosphate. As Alpl-null mice die before weaning, we aimed to generate mouse models of late-onset HPP with extended life spans by engineering a floxed Alpl allele, allowing for conditional gene ablation (conditional knockout [cKO]) when crossed with Cre recombinase transgenic mice. The authors hypothesized that targeted deletion of Alpl in osteoblasts and selected dental cells ( Col1a1-cKO) or deletion in chondrocytes, osteoblasts, and craniofacial mesenchyme ( Prx1-cKO) would phenocopy skeletal and dental manifestations of late-onset HPP. Col1a1-cKO and Prx1-cKO mice were viable and fertile, and they did not manifest the epileptic seizures characteristic of the Alpl-/- model of severe infantile HPP. Both cKO models featured normal postnatal body weight but significant reduction as compared with wild type mice by 8 to 12 wk. Plasma alkaline phosphatase for both cKO models at 24 wk was reduced by approximately 75% as compared with controls. Radiography revealed profound skeletal defects in cKO mice, including rachitic changes, hypomineralized long bones, deformations, and signs of fractures. Microcomputed tomography confirmed quantitative differences in cortical and trabecular bone, including decreased cortical thickness and mineral density. Col1a1-cKO mice exhibited classic signs of HPP dentoalveolar disease, including short molar roots with thin dentin, lack of acellular cementum, and osteoid accumulation in alveolar bone. Prx1-cKO mice exhibited the same array of periodontal defects but featured less affected molar dentin. Both cKO models exhibited reduced alveolar bone height and 4-fold increased numbers of osteoclast-like cells versus wild type at 24 wk, consistent with HPP-associated periodontal disease. These novel models of late-onset HPP can inform on long-term skeletal and dental manifestations and will provide essential tools to further studies of etiopathologies and therapeutic interventions.
Collapse
Affiliation(s)
- B L Foster
- 1 Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - P Kuss
- 2 Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - M C Yadav
- 2 Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - T N Kolli
- 1 Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - S Narisawa
- 2 Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - L Lukashova
- 3 Hospital for Special Surgery, New York, NY, USA
| | - E Cory
- 4 Department of Bioengineering, University of California-San Diego, La Jolla, CA, USA.,5 Center for Musculoskeletal Research, University of California-San Diego, La Jolla, CA, USA
| | - R L Sah
- 4 Department of Bioengineering, University of California-San Diego, La Jolla, CA, USA.,5 Center for Musculoskeletal Research, University of California-San Diego, La Jolla, CA, USA.,6 Department of Orthopaedic Surgery, University of California-San Diego, La Jolla, CA, USA
| | - M J Somerman
- 7 National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - J L Millán
- 2 Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
35
|
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
|
36
|
Galli S, Jimbo R, Naito Y, Berner S, Dard M, Wennerberg A. Chemically modified titanium-zirconium implants in comparison with commercially pure titanium controls stimulate the early molecular pathways of bone healing. Clin Oral Implants Res 2016; 28:1234-1240. [DOI: 10.1111/clr.12947] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Silvia Galli
- Department of Prosthodontics; Faculty of Odontology; Malmö University; Malmö Sweden
| | - Ryo Jimbo
- Department of Prosthodontics; Faculty of Odontology; Malmö University; Malmö Sweden
- Department of Oral and Maxillofacial Surgery and Oral Medicine; Faculty of Odontology; Malmö University; Malmö Sweden
| | - Yoshihito Naito
- Oral Implant Center; Tokushima University Hospital; Tokushima Japan
| | | | - Michel Dard
- Institut Straumann AG; Basel Switzerland
- College of Dentistry; New York University; New York NY USA
| | - Ann Wennerberg
- Department of Prosthodontics; Faculty of Odontology; Malmö University; Malmö Sweden
| |
Collapse
|
37
|
Abstract
Hypophosphatasia (HPP) is an inherited systemic bone disease that is characterized by bone hypomineralization. HPP is classified into six forms according to the age of onset and severity as perinatal (lethal), perinatal benign, infantile, childhood, adult, and odontohypophosphatasia. The causative gene of the disease is the ALPL gene that encodes tissue-nonspecific alkaline phosphatase (TNAP). TNAP is expressed ubiquitously, and its physiological role is apparent in bone mineralization. A defect in bone mineralization can manifest in several ways, including rickets or osteomalacia in HPP patients. Patients with severe forms suffer from respiratory failure because of hypoplastic chest, which is the main cause of death. They sometimes present with seizures due to a defect in vitamin B6 metabolism resulting from the lack of alkaline phosphatase activity in neuronal cells, which is also lethal. Patients with a mild form of the disease exhibit rickets or osteomalacia and a functional defect of exercise. Odontohypophosphatasia shows only dental manifestations. To date, 302 mutations in the ALPL gene have been reported, mainly single-nucleotide substitutions, and the relationships between phenotype and genotype have been partially elucidated. An established treatment for HPP was not available until the recent development of enzyme replacement therapy. The first successful enzyme replacement therapy in model mice using a modified human TNAP protein (asfotase alfa) was reported in 2008, and subsequently success in patients with severe form of the disease was reported in 2012. In 2015, asfotase alfa was approved in Japan in July, followed by in the EU and Canada in August, and then by the US Food and Drug Administration in the USA in October. It is expected that therapy with asfotase alfa will drastically change treatments and prognosis of HPP.
Collapse
Affiliation(s)
- Hideo Orimo
- Division of Metabolism and Nutrition, Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
38
|
Abstract
Hypophosphatasia is the inborn error of metabolism characterized by low serum alkaline phosphatase activity (hypophosphatasaemia). This biochemical hallmark reflects loss-of-function mutations within the gene that encodes the tissue-nonspecific isoenzyme of alkaline phosphatase (TNSALP). TNSALP is a cell-surface homodimeric phosphohydrolase that is richly expressed in the skeleton, liver, kidney and developing teeth. In hypophosphatasia, extracellular accumulation of TNSALP natural substrates includes inorganic pyrophosphate, an inhibitor of mineralization, which explains the dento-osseous and arthritic complications featuring tooth loss, rickets or osteomalacia, and calcific arthopathies. Severely affected infants sometimes also have hypercalcaemia and hyperphosphataemia due to the blocked entry of minerals into the skeleton, and pyridoxine-dependent seizures from insufficient extracellular hydrolysis of pyridoxal 5'-phosphate, the major circulating form of vitamin B6, required for neurotransmitter synthesis. Autosomal recessive or dominant inheritance from ~300 predominantly missense ALPL (also known as TNSALP) mutations largely accounts for the remarkably broad-ranging expressivity of hypophosphatasia. High serum concentrations of pyridoxal 5'-phosphate represent a sensitive and specific biochemical marker for hypophosphatasia. Also, phosphoethanolamine levels are usually elevated in serum and urine, though less reliably for diagnosis. TNSALP mutation detection is important for recurrence risk assessment and prenatal diagnosis. Diagnosing paediatric hypophosphatasia is aided by pathognomic radiographic changes when the skeletal disease is severe. Hypophosphatasia was the last type of rickets or osteomalacia to await a medical treatment. Now, significant successes for severely affected paediatric patients are recognized using asfotase alfa, a bone-targeted recombinant TNSALP.
Collapse
Affiliation(s)
- Michael P Whyte
- Center for Metabolic Bone Disease and Molecular Research, Shriners Hospital for Children, 4400 Clayton Avenue, Saint Louis, Missouri 63110, USA
- Division of Bone and Mineral Diseases, Department of Internal Medicine, Washington University School of Medicine at Barnes-Jewish Hospital, 660 South Euclid Avenue, Saint Louis, Missouri 63110, USA
| |
Collapse
|
39
|
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
|
40
|
Zweifler LE, Ao M, Yadav M, Kuss P, Narisawa S, Kolli TN, Wimer HF, Farquharson C, Somerman MJ, Millán JL, Foster BL. Role of PHOSPHO1 in Periodontal Development and Function. J Dent Res 2016; 95:742-51. [PMID: 27016531 DOI: 10.1177/0022034516640246] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The tooth root and periodontal apparatus, including the acellular and cellular cementum, periodontal ligament (PDL), and alveolar bone, are critical for tooth function. Cementum and bone mineralization is regulated by factors including enzymes and extracellular matrix proteins that promote or inhibit hydroxyapatite crystal growth. Orphan Phosphatase 1 (Phospho1, PHOSPHO1) is a phosphatase expressed by chondrocytes, osteoblasts, and odontoblasts that functions in skeletal and dentin mineralization by initiating deposition of hydroxyapatite inside membrane-limited matrix vesicles. The role of PHOSPHO1 in periodontal formation remains unknown and we aimed to determine its functional importance in these tissues. We hypothesized that the enzyme would regulate proper mineralization of the periodontal apparatus. Spatiotemporal expression of PHOSPHO1 was mapped during periodontal development, and Phospho1(-/-) mice were analyzed using histology, immunohistochemistry, in situ hybridization, radiography, and micro-computed tomography. The Phospho1 gene and PHOSPHO1 protein were expressed by active alveolar bone osteoblasts and cementoblasts during cellular cementum formation. In Phospho1(-/-) mice, acellular cementum formation and mineralization were unaffected, whereas cellular cementum deposition increased although it displayed delayed mineralization and cementoid. Phospho1(-/-) mice featured disturbances in alveolar bone mineralization, shown by accumulation of unmineralized osteoid matrix and interglobular patterns of protein deposition. Parallel to other skeletal sites, deposition of mineral-regulating protein osteopontin (OPN) was increased in alveolar bone in Phospho1(-/-) mice. In contrast to the skeleton, genetic ablation of Spp1, the gene encoding OPN, did not ameliorate dentoalveolar defects in Phospho1(-/-) mice. Despite alveolar bone mineralization defects, periodontal attachment and function appeared undisturbed in Phospho1(-/-) mice, with normal PDL architecture and no evidence of bone loss over time. This study highlights the role of PHOSPHO1 in mineralization of alveolar bone and cellular cementum, further revealing that acellular cementum formation is not substantially regulated by PHOSPHO1 and likely does not rely on matrix vesicle-mediated initiation of mineralization.
Collapse
Affiliation(s)
- L E Zweifler
- Division of Biosciences, College of Dentistry, Ohio State University, Columbus, OH, USA
| | - M Ao
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M Yadav
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - P Kuss
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - S Narisawa
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - T N Kolli
- Division of Biosciences, College of Dentistry, Ohio State University, Columbus, OH, USA
| | - H F Wimer
- Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - C Farquharson
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, UK
| | - 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 Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - B L Foster
- Division of Biosciences, College of Dentistry, Ohio State University, Columbus, OH, USA
| |
Collapse
|
41
|
Iijima O, Miyake K, Watanabe A, Miyake N, Igarashi T, Kanokoda C, Nakamura-Takahashi A, Kinoshita H, Noguchi T, Abe S, Narisawa S, Millán JL, Okada T, Shimada T. Prevention of Lethal Murine Hypophosphatasia by Neonatal Ex Vivo Gene Therapy Using Lentivirally Transduced Bone Marrow Cells. Hum Gene Ther 2015; 26:801-12. [PMID: 26467745 DOI: 10.1089/hum.2015.078] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Hypophosphatasia (HPP) is an inherited skeletal and dental disease caused by loss-of-function mutations in the gene that encodes tissue-nonspecific alkaline phosphatase (TNALP). The major symptoms of severe forms of the disease are bone defects, respiratory insufficiency, and epileptic seizures. In 2015, enzyme replacement therapy (ERT) using recombinant bone-targeted TNALP with deca-aspartate (D10) motif was approved to treat pediatric HPP patients in Japan, Canada, and Europe. However, the ERT requires repeated subcutaneous administration of the enzyme because of the short half-life in serum. In the present study, we evaluated the feasibility of neonatal ex vivo gene therapy in TNALP knockout (Akp2(-/-)) HPP mice using lentivirally transduced bone marrow cells (BMC) expressing bone-targeted TNALP in which a D10 sequence was linked to the C-terminus of soluble TNALP (TNALP-D10). The Akp2(-/-) mice usually die within 20 days because of growth failure, epileptic seizures, and hypomineralization. However, an intravenous transplantation of BMC expressing TNALP-D10 (ALP-BMC) into neonatal Akp2(-/-) mice prolonged survival of the mice with improved bone mineralization compared with untransduced BMC-transplanted Akp2(-/-) mice. The treated Akp2(-/-) mice were normal in appearance and experienced no seizures during the experimental period. The lentivirally transduced BMC were efficiently engrafted in the recipient mice and supplied TNALP-D10 continuously at a therapeutic level for at least 3 months. Moreover, TNALP-D10 overexpression did not affect multilineage reconstitution in the recipient mice. The plasma ALP activity was sustained at high levels in the treated mice, and tissue ALP activity was selectively detected on bone surfaces, not in the kidneys or other organs. No ectopic calcification was observed in the ALP-BMC-treated mice. These results indicate that lentivirally transduced BMC can serve as a reservoir for stem cell-based ERT to rescue the Akp2(-/-) phenotype. Neonatal ex vivo gene therapy thus appears to be a possible treatment option for treating severe HPP.
Collapse
Affiliation(s)
- Osamu Iijima
- 1 Division of Gene Therapy, Department of Biochemistry and Molecular Biology, Research Center for Advanced Medical Technology, Nippon Medical School, Tokyo, Japan
| | - Koichi Miyake
- 1 Division of Gene Therapy, Department of Biochemistry and Molecular Biology, Research Center for Advanced Medical Technology, Nippon Medical School, Tokyo, Japan
| | - Atsushi Watanabe
- 1 Division of Gene Therapy, Department of Biochemistry and Molecular Biology, Research Center for Advanced Medical Technology, Nippon Medical School, Tokyo, Japan .,2 Division of Clinical Genetics, Nippon Medical School Hospital, Tokyo, Japan
| | - Noriko Miyake
- 1 Division of Gene Therapy, Department of Biochemistry and Molecular Biology, Research Center for Advanced Medical Technology, Nippon Medical School, Tokyo, Japan
| | - Tsutomu Igarashi
- 1 Division of Gene Therapy, Department of Biochemistry and Molecular Biology, Research Center for Advanced Medical Technology, Nippon Medical School, Tokyo, Japan .,3 Department of Ophthalmology, Nippon Medical School Hospital, Tokyo, Japan
| | - Chizu Kanokoda
- 1 Division of Gene Therapy, Department of Biochemistry and Molecular Biology, Research Center for Advanced Medical Technology, Nippon Medical School, Tokyo, Japan
| | - Aki Nakamura-Takahashi
- 1 Division of Gene Therapy, Department of Biochemistry and Molecular Biology, Research Center for Advanced Medical Technology, Nippon Medical School, Tokyo, Japan
| | - Hideaki Kinoshita
- 4 Department of Dental Materials Science, Tokyo Dental College, Tokyo, Japan
| | - Taku Noguchi
- 5 Department of Anatomy, Tokyo Dental College, Tokyo, Japan
| | - Shinichi Abe
- 5 Department of Anatomy, Tokyo Dental College, Tokyo, Japan
| | - Sonoko Narisawa
- 6 Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute , La Jolla, California
| | - José Luis Millán
- 6 Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute , La Jolla, California
| | - Takashi Okada
- 1 Division of Gene Therapy, Department of Biochemistry and Molecular Biology, Research Center for Advanced Medical Technology, Nippon Medical School, Tokyo, Japan
| | - Takashi Shimada
- 1 Division of Gene Therapy, Department of Biochemistry and Molecular Biology, Research Center for Advanced Medical Technology, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
42
|
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
|