151
|
Zhang M, Wang H, Zhang J, Zhang H, Yang H, Wan X, Jing L, Lu L, Liu X, Yu S, Chang W, Wang M. Unilateral anterior crossbite induces aberrant mineral deposition in degenerative temporomandibular cartilage in rats. Osteoarthritis Cartilage 2016; 24:921-31. [PMID: 26746151 PMCID: PMC5699887 DOI: 10.1016/j.joca.2015.12.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 12/04/2015] [Accepted: 12/20/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To investigate whether mechanical stress induces mineral deposits that contribute to matrix degradation at the onset of osteoarthritis (OA) in temporomandibular joint (TMJ) cartilage. DESIGN Female Spraguee-Dawley rats were subjected to an unilateral anterior crossbite (UAC) procedure. Histology, electron microscopy, and energy dispersive spectrometer (EDS) were used to examine cartilage matrix structures and composition of mineral deposit in the affected TMJ cartilage. Protein and/or RNA expression of phenotypic markers and mineralization modulators and matrix degradation was analyzed by immunohistochemistry and/or real-time PCR. Synthetic basic calcium phosphate (BCP) and calcium pyrophosphate dehydrate (CPPD) crystals were used to stimulate ATDC5 cells for their impact on cell differentiation and gene expression. RESULTS Fragmented and disorganized collagen fibers, expanded fibrous spaces, and enhancement of matrix vesicle production and mineral deposition were observed in matrices surrounding hypertrophic chondrocytes in cartilage as early as 2-weeks post-UAC and exacerbated with time. The mineral deposits in TMJ cartilage at 12- and 20-weeks post-UAC had Ca/P ratios of 1.42 and 1.44, which are similar to the ratios for BCP. The expression of mineralization inhibitors, NPP1, ANK, CD73, and Matrix gla protein (MGP) was decreased from 2 to 8 weeks post-UAC, so were the chondrogenic markers, Col-2, Col-X and aggrecan. In contrast, the expression of tissue-nonspecific alkaline phosphatase (TNAP) and MMP13 was increased 4-weeks post-UAC. Treating ADTC5 cells with BCP crystals increased MMPs and ADAMTS5 expression, but reduced matrix production in a time-dependent manner. CONCLUSION UAC induces deposition of BCP-like minerals in osteoarthritic cartilage, which can stimulate matrix degradation by promoting the expression of cartilage-degrading enzymes to facilitate OA progression.
Collapse
Affiliation(s)
- M. Zhang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - H. Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - J. Zhang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - H. Zhang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - H. Yang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - X. Wan
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - L. Jing
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - L. Lu
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - X. Liu
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - S. Yu
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - W. Chang
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, USA,Department of Medicine, University of California San Francisco, USA
| | - M. Wang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, China,Address correspondence and reprint requests to: M. Wang, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China. (M. Wang)
| |
Collapse
|
152
|
Ruiz JL, Weinbaum S, Aikawa E, Hutcheson JD. Zooming in on the genesis of atherosclerotic plaque microcalcifications. J Physiol 2016; 594:2915-27. [PMID: 27040360 DOI: 10.1113/jp271339] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 03/23/2016] [Indexed: 01/08/2023] Open
Abstract
Epidemiological evidence conclusively demonstrates that calcium burden is a significant predictor of cardiovascular morbidity and mortality; however, the underlying mechanisms remain largely unknown. These observations have challenged the previously held notion that calcification serves to stabilize the atherosclerotic plaque. Recent studies have shown that microcalcifications that form within the fibrous cap of the plaques lead to the accrual of plaque-destabilizing mechanical stress. Given the association between calcification morphology and cardiovascular outcomes, it is important to understand the mechanisms leading to calcific mineral deposition and growth from the earliest stages. We highlight the open questions in the field of cardiovascular calcification and include a review of the proposed mechanisms involved in extracellular vesicle-mediated mineral deposition.
Collapse
Affiliation(s)
- Jessica L Ruiz
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sheldon Weinbaum
- Department of Biomedical Engineering, City College of New York, New York, NY, USA
| | - Elena Aikawa
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joshua D Hutcheson
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
153
|
Kerschnitzki M, Akiva A, Ben Shoham A, Asscher Y, Wagermaier W, Fratzl P, Addadi L, Weiner S. Bone mineralization pathways during the rapid growth of embryonic chicken long bones. J Struct Biol 2016; 195:82-92. [PMID: 27108185 DOI: 10.1016/j.jsb.2016.04.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/13/2016] [Accepted: 04/20/2016] [Indexed: 02/05/2023]
Abstract
The uptake and transport of ions from the environment to the site of bone formation is only partially understood and, for the most part, based on disparate observations in different animals. Here we study different aspects of the biomineralization pathways in one system, the rapidly forming long bones of the chicken embryo. We mainly used cryo-fixation and cryo-electron imaging to preserve the often unstable mineral phases in the tissues. We show the presence of surprisingly large amounts of mineral particles located inside membrane-delineated vesicles in the bone forming tissue between the blood vessels and the forming bone surface. Some of these particles are also located inside mitochondrial networks. The surfaces of the forming bones in the extracellular space contain abundant aggregates of amorphous calcium phosphate particles, but these are not enveloped by vesicle membranes. In the bone resorbing region, osteoclasts also contain many particles in both mitochondrial networks and within vesicles. Some of these particles are present also between cells. These observations, together with the previously reported observation that CaP mineral particles inside membranes are present in blood vessels, leads us to the conclusion that important components of the bone mineralization pathways in rapidly forming chicken bone are dense phase mineral particles bound within membranes. It remains to be determined whether these mineral particles are transported to the site of bone formation in the solid state, fluid state or dissolve and re-precipitate.
Collapse
Affiliation(s)
- Michael Kerschnitzki
- Department of Structural Biology, Weizmann Institute of Science, 76100 Rehovot, Israel.
| | - Anat Akiva
- Department of Structural Biology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Adi Ben Shoham
- Department of Molecular Genetics, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Yotam Asscher
- Department of Structural Biology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Wolfgang Wagermaier
- Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, Research Campus Golm, 14424 Potsdam, Germany
| | - Peter Fratzl
- Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, Research Campus Golm, 14424 Potsdam, Germany
| | - Lia Addadi
- Department of Structural Biology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Steve Weiner
- Department of Structural Biology, Weizmann Institute of Science, 76100 Rehovot, Israel
| |
Collapse
|
154
|
Orriss IR, Arnett TR, Russell RGG. Pyrophosphate: a key inhibitor of mineralisation. Curr Opin Pharmacol 2016; 28:57-68. [PMID: 27061894 DOI: 10.1016/j.coph.2016.03.003] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 03/18/2016] [Accepted: 03/24/2016] [Indexed: 12/20/2022]
Abstract
Inorganic pyrophosphate has long been known as a by-product of many intracellular biosynthetic reactions, and was first identified as a key endogenous inhibitor of biomineralisation in the 1960s. The major source of pyrophosphate appears to be extracellular ATP, which is released from cells in a controlled manner. Once released, ATP can be rapidly hydrolysed by ecto-nucleotide pyrophosphatase/phosphodiesterases to produce pyrophosphate. The main action of pyrophosphate is to directly inhibit hydroxyapatite formation thereby acting as a physiological 'water-softener'. Evidence suggests pyrophosphate may also act as a signalling molecule to influence gene expression and regulate its own production and breakdown. This review will summarise our current understanding of pyrophosphate metabolism and how it regulates bone mineralisation and prevents harmful soft tissue calcification.
Collapse
Affiliation(s)
- Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK.
| | - Timothy R Arnett
- Department of Cell and Developmental Biology, University College London, London, UK
| | - R Graham G Russell
- The Botnar Research Centre, Nuffield Orthopaedic Centre, Oxford, UK; The Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
| |
Collapse
|
155
|
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
|
156
|
Murali SK, Andrukhova O, Clinkenbeard EL, White KE, Erben RG. Excessive Osteocytic Fgf23 Secretion Contributes to Pyrophosphate Accumulation and Mineralization Defect in Hyp Mice. PLoS Biol 2016; 14:e1002427. [PMID: 27035636 PMCID: PMC4818020 DOI: 10.1371/journal.pbio.1002427] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 03/08/2016] [Indexed: 11/24/2022] Open
Abstract
X-linked hypophosphatemia (XLH) is the most frequent form of inherited rickets in humans caused by mutations in the phosphate-regulating gene with homologies to endopeptidases on the X-chromosome (PHEX). Hyp mice, a murine homologue of XLH, are characterized by hypophosphatemia, inappropriately low serum vitamin D levels, increased serum fibroblast growth factor-23 (Fgf23), and osteomalacia. Although Fgf23 is known to be responsible for hypophosphatemia and reduced vitamin D hormone levels in Hyp mice, its putative role as an auto-/paracrine osteomalacia-causing factor has not been explored. We recently reported that Fgf23 is a suppressor of tissue nonspecific alkaline phosphatase (Tnap) transcription via FGF receptor-3 (FGFR3) signaling, leading to inhibition of mineralization through accumulation of the TNAP substrate pyrophosphate. Here, we report that the pyrophosphate concentration is increased in Hyp bones, and that Tnap expression is decreased in Hyp-derived osteocyte-like cells but not in Hyp-derived osteoblasts ex vivo and in vitro. In situ mRNA expression profiling in bone cryosections revealed a ~70-fold up-regulation of Fgfr3 mRNA in osteocytes versus osteoblasts of Hyp mice. In addition, we show that blocking of increased Fgf23-FGFR3 signaling with anti-Fgf23 antibodies or an FGFR3 inhibitor partially restored the suppression of Tnap expression, phosphate production, and mineralization, and decreased pyrophosphate concentration in Hyp-derived osteocyte-like cells in vitro. In vivo, bone-specific deletion of Fgf23 in Hyp mice rescued the suppressed TNAP activity in osteocytes of Hyp mice. Moreover, treatment of wild-type osteoblasts or mice with recombinant FGF23 suppressed Tnap mRNA expression and increased pyrophosphate concentrations in the culture medium and in bone, respectively. In conclusion, we found that the cell autonomous increase in Fgf23 secretion in Hyp osteocytes drives the accumulation of pyrophosphate through auto-/paracrine suppression of TNAP. Hence, we have identified a novel mechanism contributing to the mineralization defect in Hyp mice. A novel mechanism involving autocrine and paracrine actions of fibroblast growth factor-23 contributes to the mineralization defect observed in Hyp, a mouse model for X-linked hypophosphatemia. X-linked hypophosphatemia (XLH) is the most frequent form of inherited rickets in humans. A mouse model of XLH, known as Hyp, is characterized by exceptionally low serum phosphate and vitamin D levels, increased serum levels of the hormone fibroblast growth factor-23 (Fgf23), and impaired bone mineralization. Fgf23 is secreted from two classes of bone cells known as osteoblasts and osteocytes. Fgf23 increases urinary phosphate excretion and suppresses vitamin D hormone production in the kidney. Although Fgf23 is known to be responsible for lower blood phosphate and vitamin D hormone levels in Hyp mice, its putative role as a signaling factor causing impaired mineralization has not been explored. We recently reported that Fgf23 is a suppressor of tissue nonspecific alkaline phosphatase (Tnap) gene expression via FGF receptor-3 (FGFR3) signaling in osteoblasts, leading to inhibition of mineralization through accumulation of the TNAP substrate pyrophosphate. Pyrophosphate is a potent inhibitor of mineralization. Using a combination of cell culture and animal models, we report that the increase in osteocyte Fgf23 secretion of Hyp mice leads to FGFR3-mediated suppression of TNAP with subsequent accumulation of pyrophosphate. Hence, we have identified a novel signaling mechanism by which excessive osteocytic secretion of Fgf23 contributes to the mineralization defect in Hyp mice.
Collapse
Affiliation(s)
- Sathish K. Murali
- Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Olena Andrukhova
- Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Erica L. Clinkenbeard
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Kenneth E. White
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Reinhold G. Erben
- Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
- * E-mail:
| |
Collapse
|
157
|
Li C, Cui Y, Luan J, Zhou X, Li H, Wang H, Shi L, Han J. Tenascin C affects mineralization of SaOS2 osteoblast-like cells through matrix vesicles. Drug Discov Ther 2016; 10:82-7. [PMID: 26961327 DOI: 10.5582/ddt.2016.01009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Tenascin C (TNC) is an extracellular matrix glycoprotein involved in osteogenesis and bone mineralization. In a previous study, we identified TNC protein located in the matrix vesicles (MVs) of osteoblasts. MVs are determinant in the mineralization formation. Therefore, we hypothesize whether TNC can modulate osteoblast mineralization via MVs. In this study, we demonstrated that the expression level of TNC was increased with osteoblast differentiation of osteoblast-like SaOS2 cells, and down-regulation of TNC expression by siRNA could significantly inhibit SaOS2 differentiation toward osteoblasts and mineralization as evidenced by decreases in ALP activity, mineralized nodule formation, calcium deposition, and down-regulation of osteogenic marker genes ALP, and COL1A1. Furthermore, we validated that TNC located in the MVs of mineralized SaOS2 cells, and that down-regulation of TNC could decrease MVs mineralization ability in vitro, and the decrease of MVs mineralization ability was not associated with annexins. In conclusion, in this study, we extended the role of TNC during osteogenesis previous progresses, and that supported TNC as an important functional MVs component in modulating osteoblast mineralization.
Collapse
Affiliation(s)
- Chengzhi Li
- School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Science
| | | | | | | | | | | | | | | |
Collapse
|
158
|
Kerschnitzki M, Akiva A, Shoham AB, Koifman N, Shimoni E, Rechav K, Arraf AA, Schultheiss TM, Talmon Y, Zelzer E, Weiner S, Addadi L. Transport of membrane-bound mineral particles in blood vessels during chicken embryonic bone development. Bone 2016; 83:65-72. [PMID: 26481471 DOI: 10.1016/j.bone.2015.10.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 10/13/2015] [Accepted: 10/14/2015] [Indexed: 12/25/2022]
Abstract
During bone formation in embryos, large amounts of calcium and phosphate are taken up and transported to the site where solid mineral is first deposited. The initial mineral forms in vesicles inside osteoblasts and is deposited as a highly disordered calcium phosphate phase. The mineral is then translocated to the extracellular space where it penetrates the collagen matrix and crystallizes. To date little is known about the transport mechanisms of calcium and phosphate in the vascular system, especially when high transport rates are needed and the concentrations of these ions in the blood serum may exceed the solubility product of the mineral phase. Here we used a rapidly growing biological model, the chick embryo, to study the bone mineralization pathway taking advantage of the fact that large amounts of bone mineral constituents are transported. Cryo scanning electron microscopy together with cryo energy dispersive X-ray spectroscopy and focused-ion beam imaging in the serial surface view mode surprisingly reveal the presence of abundant vesicles containing small mineral particles in the lumen of the blood vessels. Morphologically similar vesicles are also found in the cells associated with bone formation. This observation directly implicates the vascular system in solid mineral distribution, as opposed to the transport of ions in solution. Mineral particle transport inside vesicles implies that far larger amounts of the bone mineral constituents can be transported through the vasculature, without the danger of ectopic precipitation. This introduces a new stage into the bone mineral formation pathway, with the first mineral being formed far from the bone itself.
Collapse
Affiliation(s)
- Michael Kerschnitzki
- Department of Structural Biology, Weizmann Institute of Science, 76100 Rehovot, Israel.
| | - Anat Akiva
- Department of Structural Biology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Adi Ben Shoham
- Department of Molecular Genetics, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Naama Koifman
- Department of Chemical Engineering and the Russell Berrie Nanotechnology Institute (RBNI), Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Eyal Shimoni
- Department of Chemical Research Support, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Katya Rechav
- Department of Chemical Research Support, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Alaa A Arraf
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | - Thomas M Schultheiss
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 32000, Haifa, Israel
| | - Yeshayahu Talmon
- Department of Chemical Engineering and the Russell Berrie Nanotechnology Institute (RBNI), Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Stephen Weiner
- Department of Structural Biology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Lia Addadi
- Department of Structural Biology, Weizmann Institute of Science, 76100 Rehovot, Israel
| |
Collapse
|
159
|
Abstract
Hypophosphatasia (HPP) is due to deficient activity of the tissue-nonspecific isoenzyme of alkaline phosphatase (TNAP). This enzyme cleaves extracellular substrates inorganic pyrophosphates (PPi), pyridoxal-5'-phosphate (PLP), phosphoethanolamine (PEA) and nucleotides, and probably other substrates not yet identified. During the last 15 years the role of TNAP in mineralization, and to a less degree in brain, has been investigated, providing hypotheses and explanations for both bone and neuronal HPP phenotypes. ALPL, the gene encoding TNAP, is subject to many mutations, mostly missense mutations. A few number of mutations are recurrently found and may be quite frequent in particular populations. This reflects founder effects. The great variety of mutations results in a great number of compound heterozygous genotypes and in highly variable clinical expressivity. A good correlation was observed between the severity of the disease and in vitro enzymatic activity of the mutant protein measured after site-directed mutagenesis. Many missense mutations found in severe hypophosphatasia produced a mutant protein that failed to reach the cell membrane , was accumulated in the cis-Golgi and was subsequently degraded in the proteasome. Missense mutations located in the catalytic site or in the homodimer interface were often shown by site-directed mutagenesis to have a dominant negative effect. Currently molecular diagnosis of HPP is based on the sequencing of the coding sequence of ALPL that allows detection of approximately 95 % of mutations in severe cases. In addition, other genes, especially genes encoding proteins involved in the regulation of extracellular PPi concentration, could modify the phenotype (modifier genes).
Collapse
|
160
|
Cianciolo G, Capelli I, Cappuccilli M, Schillaci R, Cozzolino M, La Manna G. Calcifying circulating cells: an uncharted area in the setting of vascular calcification in CKD patients. Clin Kidney J 2016; 9:280-6. [PMID: 26985381 PMCID: PMC4792620 DOI: 10.1093/ckj/sfv145] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 11/27/2015] [Indexed: 12/16/2022] Open
Abstract
Vascular calcification, occurring during late-stage vascular and valvular disease, is highly associated with chronic kidney disease-mineral and bone disorders (CKD-MBD), representing a major risk factor for cardiovascular morbidity and mortality. The hallmark of vascular calcification, which involves both media and intima, is represented by the activation of cells committed to an osteogenic programme. Several studies have analysed the role of circulating calcifying cells (CCCs) in vascular calcification. CCCs are bone marrow (BM)-derived cells with an osteogenic phenotype, participating in intima calcification processes and defined by osteocalcin and bone alkaline phosphatase expression. The identification of CCCs in diabetes and atherosclerosis is the most recent, intriguing and yet uncharted chapter in the scenario of the bone-vascular axis. Whether osteogenic shift occurs in the BM, the bloodstream or both, is not known, and also the factors promoting CCC formation have not been identified. However, it is possible to recognize a common pathogenic commitment of inflammation in atherosclerosis and diabetes, in which metabolic control may also have a role. Currently available studies in patients without CKD did not find an association of CCCs with markers of bone metabolism. Preliminary data on CKD patients indicate an implication of mineral bone disease in vascular calcification, as a consequence of functional and anatomic integrity interruption of BM niches. Given the pivotal role that parathyroid hormone and osteoblasts play in regulating expansion, mobilization and homing of haematopoietic stem/progenitors cells, CKD-MBD could promote CCC formation.
Collapse
Affiliation(s)
- Giuseppe Cianciolo
- Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, Department of Experimental Diagnostic and Specialty Medicine (DIMES) , University of Bologna , Bologna , Italy
| | - Irene Capelli
- Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, Department of Experimental Diagnostic and Specialty Medicine (DIMES) , University of Bologna , Bologna , Italy
| | - Maria Cappuccilli
- Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, Department of Experimental Diagnostic and Specialty Medicine (DIMES) , University of Bologna , Bologna , Italy
| | - Roberto Schillaci
- Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, Department of Experimental Diagnostic and Specialty Medicine (DIMES) , University of Bologna , Bologna , Italy
| | - Mario Cozzolino
- Nephrology and Dialysis, S. Paolo Hospital, Department of Health Sciences (DISS) , University of Milan , Milan , Italy
| | - Gaetano La Manna
- Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, Department of Experimental Diagnostic and Specialty Medicine (DIMES) , University of Bologna , Bologna , Italy
| |
Collapse
|
161
|
Murali SK, Roschger P, Zeitz U, Klaushofer K, Andrukhova O, Erben RG. FGF23 Regulates Bone Mineralization in a 1,25(OH)2 D3 and Klotho-Independent Manner. J Bone Miner Res 2016; 31:129-42. [PMID: 26235988 DOI: 10.1002/jbmr.2606] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 07/23/2015] [Accepted: 07/24/2015] [Indexed: 12/25/2022]
Abstract
Fibroblast growth factor-23 (Fgf23) is a bone-derived hormone, suppressing phosphate reabsorption and vitamin D hormone (1,25(OH)2 D3 ) production in the kidney. It has long been an enigma why lack of Fgf23 or of Klotho, the coreceptor for Fgf23, leads to severe impairment in bone mineralization despite the presence of hypercalcemia and hyperphosphatemia. Using Fgf23(-/-) or Klotho(-/-) mice together with compound mutant mice lacking both Fgf23 or Klotho and a functioning vitamin D receptor, we show that in Klotho(-/-) mice the mineralization defect is solely driven by 1,25(OH)2 D3 -induced upregulation of the mineralization-inhibiting molecules osteopontin and pyrophosphate in bone. In Fgf23(-/-) mice, the mineralization defect has two components, a 1,25(OH)2 D3 -driven component similar to Klotho(-/-) mice and a component driven by lack of Fgf23, causing additional accumulation of osteopontin. We found that FGF23 regulates osteopontin secretion indirectly by suppressing alkaline phosphatase transcription and phosphate production in osteoblastic cells, acting through FGF receptor-3 in a Klotho-independent manner. Hence, FGF23 secreted from osteocytes may form an autocrine/paracrine feedback loop for the local fine-tuning of bone mineralization.
Collapse
Affiliation(s)
- Sathish Kumar Murali
- Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Paul Roschger
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of WGKK and AUVA Trauma Center Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria
| | - Ute Zeitz
- Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Klaus Klaushofer
- Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of WGKK and AUVA Trauma Center Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria
| | - Olena Andrukhova
- Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Reinhold G Erben
- Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
162
|
Javaheri B, Carriero A, Staines KA, Chang YM, Houston DA, Oldknow KJ, Millan JL, Kazeruni BN, Salmon P, Shefelbine S, Farquharson C, Pitsillides AA. Phospho1 deficiency transiently modifies bone architecture yet produces consistent modification in osteocyte differentiation and vascular porosity with ageing. Bone 2015; 81:277-291. [PMID: 26232374 PMCID: PMC4652607 DOI: 10.1016/j.bone.2015.07.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 07/24/2015] [Accepted: 07/27/2015] [Indexed: 12/11/2022]
Abstract
PHOSPHO1 is one of principal proteins involved in initiating bone matrix mineralisation. Recent studies have found that Phospho1 KO mice (Phospho1-R74X) display multiple skeletal abnormalities with spontaneous fractures, bowed long bones, osteomalacia and scoliosis. These analyses have however been limited to young mice and it remains unclear whether the role of PHOSPHO1 is conserved in the mature murine skeleton where bone turnover is limited. In this study, we have used ex-vivo computerised tomography to examine the effect of Phospho1 deletion on tibial bone architecture in mice at a range of ages (5, 7, 16 and 34 weeks of age) to establish whether its role is conserved during skeletal growth and maturation. Matrix mineralisation has also been reported to influence terminal osteoblast differentiation into osteocytes and we have also explored whether hypomineralised bones in Phospho1 KO mice exhibit modified osteocyte lacunar and vascular porosity. Our data reveal that Phospho1 deficiency generates age-related defects in trabecular architecture and compromised cortical microarchitecture with greater porosity accompanied by marked alterations in osteocyte shape, significant increases in osteocytic lacuna and vessel number. Our in vitro studies examining the behaviour of osteoblast derived from Phospho1 KO and wild-type mice reveal reduced levels of matrix mineralisation and modified osteocytogenic programming in cells deficient in PHOSPHO1. Together our data suggest that deficiency in PHOSPHO1 exerts modifications in bone architecture that are transient and depend upon age, yet produces consistent modification in lacunar and vascular porosity. It is possible that the inhibitory role of PHOSPHO1 on osteocyte differentiation leads to these age-related changes in bone architecture. It is also intriguing to note that this apparent acceleration in osteocyte differentiation evident in the hypomineralised bones of Phospho1 KO mice suggests an uncoupling of the interplay between osteocytogenesis and biomineralisation. Further studies are required to dissect the molecular processes underlying the regulatory influences exerted by PHOSPHO1 on the skeleton with ageing.
Collapse
Affiliation(s)
- B Javaheri
- The Royal Veterinary College, London, United Kingdom.
| | - A Carriero
- Department of Biomedical Engineering, Florida Institute of Technology Melbourne, FL 32901, USA
| | - K A Staines
- The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, United Kingdom
| | - Y-M Chang
- The Royal Veterinary College, London, United Kingdom
| | - D A Houston
- The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, United Kingdom
| | - K J Oldknow
- The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, United Kingdom
| | - J L Millan
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | | | - P Salmon
- Bruker-microCT, Kartuizersweg 3B, 2550 Kontich, Belgium
| | - S Shefelbine
- Department of Mechanical and Industrial Engineering, Northeastern University, USA
| | - C Farquharson
- The Roslin Institute and R(D)SVS, University of Edinburgh, Edinburgh, United Kingdom
| | | |
Collapse
|
163
|
Nacre extract restores the mineralization capacity of subchondral osteoarthritis osteoblasts. J Struct Biol 2015; 192:500-509. [DOI: 10.1016/j.jsb.2015.10.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 10/15/2015] [Accepted: 10/16/2015] [Indexed: 12/16/2022]
|
164
|
Bianchi ML. Hypophosphatasia: an overview of the disease and its treatment. Osteoporos Int 2015; 26:2743-57. [PMID: 26245849 DOI: 10.1007/s00198-015-3272-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 07/28/2015] [Indexed: 11/30/2022]
Abstract
This review presents the current knowledge on hypophosphatasia, a rare genetic disease of very variable severity (from lethal to mild) and clinical presentation, caused by defective production of tissue-non-specific alkaline phosphatase (TNSALP). Hypophosphatasia can affect babies in utero as well as infants, children, and adults. The article first presents the genetics of TNSALP and its many known mutations underlying the disease. Then, it presents the epidemiology, classification, and clinical presentation of the six different forms of the disease (perinatal lethal, prenatal benign, infantile, childhood, adult, and odontohypophosphatasia) as well as the essential diagnostic clues. The last section on treatment presents a survey of the therapeutic approaches, up to the ongoing phase 2 studies of enzyme replacement therapy.
Collapse
Affiliation(s)
- M L Bianchi
- Experimental Laboratory for Children's Bone Metabolism Research, Bone Metabolism Unit, Istituto Auxologico Italiano IRCCS, via L. Ariosto 13, 20145, Milano, Italy.
| |
Collapse
|
165
|
Application of Green Tea Catechin for Inducing the Osteogenic Differentiation of Human Dedifferentiated Fat Cells in Vitro. Int J Mol Sci 2015; 16:27988-8000. [PMID: 26602917 PMCID: PMC4691028 DOI: 10.3390/ijms161226081] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 11/16/2015] [Accepted: 11/18/2015] [Indexed: 12/21/2022] Open
Abstract
Despite advances in stem cell biology, there are few effective techniques to promote the osteogenic differentiation of human primary dedifferentiated fat (DFAT) cells. We attempted to investigate whether epigallocatechin-3-gallate (EGCG), the main component of green tea catechin, facilitates early osteogenic differentiation and mineralization on DFAT cells in vitro. DFAT cells were treated with EGCG (1.25-10 μM) in osteogenic medium (OM) with or without 100 nM dexamethasone (Dex) for 12 days (hereafter two osteogenic media were designated as OM(Dex) and OM). Supplementation of 1.25 μM EGCG to both the media effectively increased the mRNA expression of collagen 1 (COL1A1) and runt-related transcription factor 2 (RUNX2) and also increased proliferation and mineralization. Compared to OM(Dex) with EGCG, OM with EGCG induced earlier expression for COL1A1 and RUNX2 at day 1 and higher mineralization level at day 12. OM(Dex) with 10 μM EGCG remarkably hampered the proliferation of the DFAT cells. These results suggest that OM(without Dex) with EGCG might be a preferable medium to promote proliferation and to induce osteoblast differentiation of DFAT cells. Our findings provide an insight for the combinatory use of EGCG and DFAT cells for bone regeneration and stem cell-based therapy.
Collapse
|
166
|
Lee HB, Kim J, Kim SH, Kim S, Kim OJ, Oh SH. Association between Serum Alkaline Phosphatase Level and Cerebral Small Vessel Disease. PLoS One 2015; 10:e0143355. [PMID: 26580067 PMCID: PMC4651565 DOI: 10.1371/journal.pone.0143355] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 11/03/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Serum alkaline phosphatase (ALP) is a marker of vascular calcification. A high serum ALP level is associated with an increase in cardiovascular events, and predicts poor functional outcome in patients with stroke. We investigated whether serum ALP was associated with cerebral small vessel disease (cSVD) and large cerebral artery stenosis (LCAS). METHODS We evaluated vascular risk factors, brain magnetic resonance images (MRIs), and MR angiograms from 1,011 neurologically healthy participants. The presence of silent lacunar infarction (SLI) and moderate-to-severe cerebral white matter hyperintensities (MS-cWMH) were evaluated as indices of cSVD on brain MRIs. Findings of extracranial arterial stenosis (ECAS) or intracranial arterial stenosis (ICAS) were considered to be indices of LCAS on MR angiograms. RESULTS Subjects with SLI (odds ratio [OR]: 2.09; 95% confidence interval [CI]: 1.27-3.42; p = 0.004) and MS-cWMH (OR: 1.48; 95% CI; 1.03-2.13, p = 0.036) were significantly more likely to have ALP levels in the third tertile (ALP ≥ 195 IU/L) than the first tertile (ALP ≤ 155 IU/L), after adjusting for cardiovascular risk factors. The mean serum ALP level was significantly higher in patients with SLI or MS-cWMH compared to patients without those findings. After adjustment for confounding factors, the multivariate model found that the statistical significance of serum ALP remained when the presence of SLI (OR: 1.05 per 10 IU/L increase in ALP; 95% CI: 1.02-1.08; p = 0.003) or MS-cWMH (OR: 1.03 per 10 IU/L increase in ALP; 95% CI: 1.00-1.06; p = 0.025) were added to the model. There were no differences in the proportions of patients with LCAS, ICAS, and ECAS across the serum ALP tertiles. CONCLUSIONS Our study of neurologically healthy participants found a positive association between serum ALP level and indicators of cSVD, but no association between serum ALP level and the indicators of LCAS.
Collapse
Affiliation(s)
- Han-Bin Lee
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Jinkwon Kim
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Sang-Heum Kim
- Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Soonhag Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, South Korea
- Catholic Kwandong University International St. Mary’s Hospital, Incheon Metropolitan City, South Korea
| | - Ok-Joon Kim
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Seung-Hun Oh
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
- * E-mail:
| |
Collapse
|
167
|
Orriss IR, Key ML, Hajjawi MOR, Millán JL, Arnett TR. Acidosis is a key regulator of osteoblast ecto-nucleotidase pyrophosphatase/phosphodiesterase 1 (NPP1) expression and activity. J Cell Physiol 2015; 230:3049-56. [PMID: 26033523 PMCID: PMC4549203 DOI: 10.1002/jcp.25041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/07/2015] [Indexed: 01/19/2023]
Abstract
Previous work has shown that acidosis prevents bone nodule formation by osteoblasts in vitro by inhibiting mineralisation of the collagenous matrix. The ratio of phosphate (Pi) to pyrophosphate (PPi) in the bone microenvironment is a fundamental regulator of bone mineralisation. Both Pi and PPi, a potent inhibitor of mineralisation, are generated from extracellular nucleotides by the actions of ecto‐nucleotidases. This study investigated the expression and activity of ecto‐nucleotidases by osteoblasts under normal and acid conditions. We found that osteoblasts express mRNA for a number of ecto‐nucleotidases including NTPdase 1–6 (ecto‐nucleoside triphosphate diphosphohydrolase) and NPP1‐3 (ecto‐nucleotide pyrophosphatase/phosphodiesterase). The rank order of mRNA expression in differentiating rat osteoblasts (day 7) was Enpp1 > NTPdase 4 > NTPdase 6 > NTPdase 5 > alkaline phosphatase > ecto‐5‐nucleotidase > Enpp3 > NTPdase 1 > NTPdase 3 > Enpp2 > NTPdase 2. Acidosis (pH 6.9) upregulated NPP1 mRNA (2.8‐fold) and protein expression at all stages of osteoblast differentiation compared to physiological pH (pH 7.4); expression of other ecto‐nucleotidases was unaffected. Furthermore, total NPP activity was increased up to 53% in osteoblasts cultured in acid conditions (P < 0.001). Release of ATP, one of the key substrates for NPP1, from osteoblasts, was unaffected by acidosis. Further studies showed that mineralised bone formation by osteoblasts cultured from NPP1 knockout mice was increased compared with wildtypes (2.5‐fold, P < 0.001) and was partially resistant to the inhibitory effect of acidosis. These results indicate that increased NPP1 expression and activity might contribute to the decreased mineralisation observed when osteoblasts are exposed to acid conditions. J. Cell. Physiol. 230: 3049–3056, 2015. © 2015 The Authors. Journal of Cellular Physiology Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK.,Department of Cell and Developmental Biology, University College London, London, UK
| | - Michelle L Key
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Mark O R Hajjawi
- Department of Cell and Developmental Biology, University College London, London, UK
| | - José L Millán
- Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Timothy R Arnett
- Department of Cell and Developmental Biology, University College London, London, UK
| |
Collapse
|
168
|
Perticone F, Perticone M, Maio R, Sciacqua A, Andreucci M, Tripepi G, Corrao S, Mallamaci F, Sesti G, Zoccali C. Serum Alkaline Phosphatase Negatively Affects Endothelium-Dependent Vasodilation in Naïve Hypertensive Patients. Hypertension 2015; 66:874-80. [DOI: 10.1161/hypertensionaha.115.06117] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Francesco Perticone
- From the Departments of Medical and Surgical Sciences (F.P., R.M., A.S., G.S.), Experimental and Clinical Medicine (M.P.), and Health Sciences (M.A.), University Magna Græcia of Catanzaro, Catanzaro, Italy; CNR-IBIM, National Research Council-Institute of Biomedicine, Reggio Calabria, Italy (G.T., F.M., C.Z.); and Biomedical Department of Internal Medicine and Subspecialties, University of Palermo, Palermo, Italy (S.C.)
| | - Maria Perticone
- From the Departments of Medical and Surgical Sciences (F.P., R.M., A.S., G.S.), Experimental and Clinical Medicine (M.P.), and Health Sciences (M.A.), University Magna Græcia of Catanzaro, Catanzaro, Italy; CNR-IBIM, National Research Council-Institute of Biomedicine, Reggio Calabria, Italy (G.T., F.M., C.Z.); and Biomedical Department of Internal Medicine and Subspecialties, University of Palermo, Palermo, Italy (S.C.)
| | - Raffaele Maio
- From the Departments of Medical and Surgical Sciences (F.P., R.M., A.S., G.S.), Experimental and Clinical Medicine (M.P.), and Health Sciences (M.A.), University Magna Græcia of Catanzaro, Catanzaro, Italy; CNR-IBIM, National Research Council-Institute of Biomedicine, Reggio Calabria, Italy (G.T., F.M., C.Z.); and Biomedical Department of Internal Medicine and Subspecialties, University of Palermo, Palermo, Italy (S.C.)
| | - Angela Sciacqua
- From the Departments of Medical and Surgical Sciences (F.P., R.M., A.S., G.S.), Experimental and Clinical Medicine (M.P.), and Health Sciences (M.A.), University Magna Græcia of Catanzaro, Catanzaro, Italy; CNR-IBIM, National Research Council-Institute of Biomedicine, Reggio Calabria, Italy (G.T., F.M., C.Z.); and Biomedical Department of Internal Medicine and Subspecialties, University of Palermo, Palermo, Italy (S.C.)
| | - Michele Andreucci
- From the Departments of Medical and Surgical Sciences (F.P., R.M., A.S., G.S.), Experimental and Clinical Medicine (M.P.), and Health Sciences (M.A.), University Magna Græcia of Catanzaro, Catanzaro, Italy; CNR-IBIM, National Research Council-Institute of Biomedicine, Reggio Calabria, Italy (G.T., F.M., C.Z.); and Biomedical Department of Internal Medicine and Subspecialties, University of Palermo, Palermo, Italy (S.C.)
| | - Giovanni Tripepi
- From the Departments of Medical and Surgical Sciences (F.P., R.M., A.S., G.S.), Experimental and Clinical Medicine (M.P.), and Health Sciences (M.A.), University Magna Græcia of Catanzaro, Catanzaro, Italy; CNR-IBIM, National Research Council-Institute of Biomedicine, Reggio Calabria, Italy (G.T., F.M., C.Z.); and Biomedical Department of Internal Medicine and Subspecialties, University of Palermo, Palermo, Italy (S.C.)
| | - Salvatore Corrao
- From the Departments of Medical and Surgical Sciences (F.P., R.M., A.S., G.S.), Experimental and Clinical Medicine (M.P.), and Health Sciences (M.A.), University Magna Græcia of Catanzaro, Catanzaro, Italy; CNR-IBIM, National Research Council-Institute of Biomedicine, Reggio Calabria, Italy (G.T., F.M., C.Z.); and Biomedical Department of Internal Medicine and Subspecialties, University of Palermo, Palermo, Italy (S.C.)
| | - Francesca Mallamaci
- From the Departments of Medical and Surgical Sciences (F.P., R.M., A.S., G.S.), Experimental and Clinical Medicine (M.P.), and Health Sciences (M.A.), University Magna Græcia of Catanzaro, Catanzaro, Italy; CNR-IBIM, National Research Council-Institute of Biomedicine, Reggio Calabria, Italy (G.T., F.M., C.Z.); and Biomedical Department of Internal Medicine and Subspecialties, University of Palermo, Palermo, Italy (S.C.)
| | - Giorgio Sesti
- From the Departments of Medical and Surgical Sciences (F.P., R.M., A.S., G.S.), Experimental and Clinical Medicine (M.P.), and Health Sciences (M.A.), University Magna Græcia of Catanzaro, Catanzaro, Italy; CNR-IBIM, National Research Council-Institute of Biomedicine, Reggio Calabria, Italy (G.T., F.M., C.Z.); and Biomedical Department of Internal Medicine and Subspecialties, University of Palermo, Palermo, Italy (S.C.)
| | - Carmine Zoccali
- From the Departments of Medical and Surgical Sciences (F.P., R.M., A.S., G.S.), Experimental and Clinical Medicine (M.P.), and Health Sciences (M.A.), University Magna Græcia of Catanzaro, Catanzaro, Italy; CNR-IBIM, National Research Council-Institute of Biomedicine, Reggio Calabria, Italy (G.T., F.M., C.Z.); and Biomedical Department of Internal Medicine and Subspecialties, University of Palermo, Palermo, Italy (S.C.)
| |
Collapse
|
169
|
Graser S, Mentrup B, Schneider D, Klein-Hitpass L, Jakob F, Hofmann C. Overexpression of tissue-nonspecific alkaline phosphatase increases the expression of neurogenic differentiation markers in the human SH-SY5Y neuroblastoma cell line. Bone 2015; 79:150-61. [PMID: 26032516 DOI: 10.1016/j.bone.2015.05.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 04/24/2015] [Accepted: 05/23/2015] [Indexed: 12/16/2022]
Abstract
Patients suffering from the rare hereditary disease hypophosphatasia (HPP), which is based on mutations in the ALPL gene, tend to develop central nervous system (CNS) related issues like epileptic seizures and neuropsychiatric illnesses such as anxiety and depression, in addition to well-known problems with the mineralization of bones and teeth. Analyses of the molecular role of tissue-nonspecific alkaline phosphatase (TNAP) in transgenic SH-SY5Y(TNAPhigh) neuroblastoma cells compared to SH-SY5Y(TNAPlow) cells indicate that the enzyme influences the expression levels of neuronal marker genes like RNA-binding protein, fox-1 homolog 3 (NEUN) and enolase 2, gamma neuronal (NSE) as well as microtubule-binding proteins like microtubule-associated protein 2 (MAP2) and microtubule-associated protein tau (TAU) during neurogenic differentiation. Fluorescence staining of SH-SY5Y(TNAPhigh) cells reveals TNAP localization throughout the whole length of the developed projection network and even synapsin Ι co-localization with strong TNAP signals at some spots at least at the early time points of differentiation. Additional immunocytochemical staining shows higher MAP2 expression in SH-SY5Y(TNAPhigh) cells and further a distinct up-regulation of tau and MAP2 in the course of neurogenic differentiation. Interestingly, transgenic SH-SY5Y(TNAPhigh) cells are able to develop longer cellular processes compared to control cells after stimulation with all-trans retinoic acid (RA). Current therapies for HPP prioritize improvement of the bone phenotype. Unraveling the molecular role of TNAP in extraosseous tissues, like in the CNS, will help to improve treatment strategies for HPP patients. Taking this rare disease as a model may also help to dissect TNAP's role in neurodegenerative diseases and even improve future treatment of common pathologies.
Collapse
Affiliation(s)
- Stephanie Graser
- Orthopedic Department, Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Germany
| | - Birgit Mentrup
- Orthopedic Department, Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Germany
| | - Doris Schneider
- Orthopedic Department, Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Germany
| | - Ludger Klein-Hitpass
- Institute of Cell Biology, Faculty of Medicine, University of Duisburg-Essen, Germany
| | - Franz Jakob
- Orthopedic Department, Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Germany
| | - Christine Hofmann
- Children's Hospital, Section of Pediatric Rheumatology and Osteology, University of Wuerzburg, Germany.
| |
Collapse
|
170
|
Bionaz M, Monaco E, Wheeler MB. Transcription Adaptation during In Vitro Adipogenesis and Osteogenesis of Porcine Mesenchymal Stem Cells: Dynamics of Pathways, Biological Processes, Up-Stream Regulators, and Gene Networks. PLoS One 2015; 10:e0137644. [PMID: 26398344 PMCID: PMC4580618 DOI: 10.1371/journal.pone.0137644] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 07/27/2015] [Indexed: 12/20/2022] Open
Abstract
The importance of mesenchymal stem cells (MSC) for bone regeneration is growing. Among MSC the bone marrow-derived stem cells (BMSC) are considered the gold standard in tissue engineering and regenerative medicine; however, the adipose-derived stem cells (ASC) have very similar properties and some advantages to be considered a good alternative to BMSC. The molecular mechanisms driving adipogenesis are relatively well-known but mechanisms driving osteogenesis are poorly known, particularly in pig. In the present study we have used transcriptome analysis to unravel pathways and biological functions driving in vitro adipogenesis and osteogenesis in BMSC and ASC. The analysis was performed using the novel Dynamic Impact Approach and functional enrichment analysis. In addition, a k-mean cluster analysis in association with enrichment analysis, networks reconstruction, and transcription factors overlapping analysis were performed in order to uncover the coordination of biological functions underlining differentiations. Analysis indicated a larger and more coordinated transcriptomic adaptation during adipogenesis compared to osteogenesis, with a larger induction of metabolism, particularly lipid synthesis (mostly triglycerides), and a larger use of amino acids for synthesis of feed-forward adipogenic compounds, larger cell signaling, lower cell-to-cell interactions, particularly for the cytoskeleton organization and cell junctions, and lower cell proliferation. The coordination of adipogenesis was mostly driven by Peroxisome Proliferator-activated Receptors together with other known adipogenic transcription factors. Only a few pathways and functions were more induced during osteogenesis compared to adipogenesis and some were more inhibited during osteogenesis, such as cholesterol and protein synthesis. Up-stream transcription factor analysis indicated activation of several lipid-related transcription regulators (e.g., PPARs and CEBPα) during adipogenesis but osteogenesis was driven by inhibition of several up-stream regulators, such as MYC. Between MSCs the data indicated an ‘adipocyte memory’ in ASC with also an apparent lower immunogenicity compared to BMSC during differentiations. Overall the analysis allowed proposing a dynamic model for the adipogenic and osteogenic differentiation in porcine ASC and BMSC.
Collapse
Affiliation(s)
- Massimo Bionaz
- Laboratory of Stem Cell Biology and Engineering in the Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Elisa Monaco
- Laboratory of Stem Cell Biology and Engineering in the Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Matthew B. Wheeler
- Laboratory of Stem Cell Biology and Engineering in the Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
171
|
Yang D, Turner AG, Wijenayaka AR, Anderson PH, Morris HA, Atkins GJ. 1,25-Dihydroxyvitamin D3 and extracellular calcium promote mineral deposition via NPP1 activity in a mature osteoblast cell line MLO-A5. Mol Cell Endocrinol 2015; 412:140-7. [PMID: 26054750 DOI: 10.1016/j.mce.2015.06.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 06/01/2015] [Accepted: 06/03/2015] [Indexed: 10/23/2022]
Abstract
While vitamin D supplementation is common, the anabolic mechanisms that improve bone status are poorly understood. Under standard mineralising conditions including media ionised calcium of 1.1 mM, 1,25-dihydroxyvitamin D3 (1,25D) enhanced differentiation and mineral deposition by the mature osteoblast/pre-osteocyte cell line, MLO-A5. This effect was markedly increased with a higher ionised calcium level (1.5 mM). Gene expression analyses revealed that 1,25D-induced mineral deposition was associated with induction of Enpp1 mRNA, coding for nucleotide pyrophosphatase phosphodiesterase 1 (NPP1) and NPP1 protein levels. Since MLO-A5 cells express abundant alkaline phosphatase that was not further modified by 1,25D treatment or exposure to increased calcium, this finding suggested that the NPP1 production of pyrophosphate (PPi) may provide alkaline phosphatase with substrate for the generation of inorganic phosphate (Pi). Consistent with this, co-treatment with Enpp1 siRNA or a NPP1 inhibitor, PPADS, abrogated 1,25D-induced mineral deposition. These data demonstrate that 1,25D stimulates osteoblast differentiation and mineral deposition, and interacts with the extracellular calcium concentration. 1,25D regulates Enpp1 expression, which presumably, in the context of adequate tissue non-specific alkaline phosphatase activity, provides Pi to stimulate mineralisation. Our findings suggest a mechanism by which vitamin D with adequate dietary calcium can improve bone mineral status.
Collapse
Affiliation(s)
- Dongqing Yang
- Bone Cell Biology Group, Centre for Orthopaedic and Trauma Research, University of Adelaide, Adelaide, SA 5005, Australia; Discipline of Medicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - Andrew G Turner
- Discipline of Medicine, University of Adelaide, Adelaide, SA 5005, Australia; Musculoskeletal Biology Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Asiri R Wijenayaka
- Bone Cell Biology Group, Centre for Orthopaedic and Trauma Research, University of Adelaide, Adelaide, SA 5005, Australia
| | - Paul H Anderson
- Discipline of Medicine, University of Adelaide, Adelaide, SA 5005, Australia; Musculoskeletal Biology Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Howard A Morris
- Discipline of Medicine, University of Adelaide, Adelaide, SA 5005, Australia; Musculoskeletal Biology Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia; Endocrine Bone Research, Chemical Pathology, SA Pathology, Adelaide, SA 5000, Australia
| | - Gerald J Atkins
- Bone Cell Biology Group, Centre for Orthopaedic and Trauma Research, University of Adelaide, Adelaide, SA 5005, Australia.
| |
Collapse
|
172
|
Ebert R, Benisch P, Krug M, Zeck S, Meißner-Weigl J, Steinert A, Rauner M, Hofbauer L, Jakob F. Acute phase serum amyloid A induces proinflammatory cytokines and mineralization via toll-like receptor 4 in mesenchymal stem cells. Stem Cell Res 2015; 15:231-9. [PMID: 26135899 DOI: 10.1016/j.scr.2015.06.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 05/20/2015] [Accepted: 06/21/2015] [Indexed: 01/05/2023] Open
Abstract
The role of serum amyloid A (SAA) proteins, which are ligands for toll-like receptors, was analyzed in human bone marrow-derived mesenchymal stem cells (hMSCs) and their osteogenic offspring with a focus on senescence, differentiation and mineralization. In vitro aged hMSC developed a senescence-associated secretory phenotype (SASP), resulting in enhanced SAA1/2, TLR2/4 and proinflammatory cytokine (IL6, IL8, IL1β, CXCL1, CXCL2) expression before entering replicative senescence. Recombinant human SAA1 (rhSAA1) induced SASP-related genes and proteins in MSC, which could be abolished by cotreatment with the TLR4-inhibitor CLI-095. The same pattern of SASP-resembling genes was stimulated upon induction of osteogenic differentiation, which is accompanied by autocrine SAA1/2 expression. In this context additional rhSAA1 enhanced the SASP-like phenotype, accelerated the proinflammatory phase of osteogenic differentiation and enhanced mineralization. Autocrine/paracrine and rhSAA1 via TLR4 stimulate a proinflammatory phenotype that is both part of the early phase of osteogenic differentiation and the development of senescence. This signaling cascade is tightly involved in bone formation and mineralization, but may also propagate pathological extraosseous calcification conditions such as calcifying inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Regina Ebert
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Brettreichstrasse 11, 97074 Würzburg, Germany.
| | - Peggy Benisch
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Brettreichstrasse 11, 97074 Würzburg, Germany
| | - Melanie Krug
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Brettreichstrasse 11, 97074 Würzburg, Germany
| | - Sabine Zeck
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Brettreichstrasse 11, 97074 Würzburg, Germany
| | - Jutta Meißner-Weigl
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Brettreichstrasse 11, 97074 Würzburg, Germany
| | - Andre Steinert
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Brettreichstrasse 11, 97074 Würzburg, Germany
| | - Martina Rauner
- Division of Endocrinology, Diabetes Bone Metabolism, Technical University of Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Lorenz Hofbauer
- Division of Endocrinology, Diabetes Bone Metabolism, Technical University of Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Brettreichstrasse 11, 97074 Würzburg, Germany.
| |
Collapse
|
173
|
Bae WJ, Jue SS, Kim SY, Moon JH, Kim EC. Effects of Sodium Tri- and Hexametaphosphate on Proliferation, Differentiation, and Angiogenic Potential of Human Dental Pulp Cells. J Endod 2015; 41:896-902. [DOI: 10.1016/j.joen.2015.01.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 01/29/2015] [Accepted: 01/30/2015] [Indexed: 01/23/2023]
|
174
|
Huesa C, Staines KA, Millán JL, MacRae VE. Effects of etidronate on the Enpp1⁻/⁻ mouse model of generalized arterial calcification of infancy. Int J Mol Med 2015; 36:159-65. [PMID: 25975272 PMCID: PMC4494596 DOI: 10.3892/ijmm.2015.2212] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/22/2015] [Indexed: 12/23/2022] Open
Abstract
Generalized arterial calcification of infancy (GACI) is an autosomal recessive disorder of spontaneous infantile arterial and periarticular calcification which is attributed to mutations in the ectonucleotide pyrophosphatase/phosphodiesterase 1 (Enpp1) gene. Whilst the bisphosphonate, etidronate, is currently used off-label for the treatment for GACI, recent studies have highlighted its detrimental effects on bone mineralisation. In the present study, we used the Enpp1-/- mouse model of GACI to examine the effects of etidronate treatment (100 µg/kg), on vascular and skeletal calcification. Micro-computed tomography (µCT) analysis revealed a significant decrease in trabecular bone mass, as reflected by the decrease in trabecular bone volume/tissue volume (BV/TV; %), trabecular thickness, trabecular separation, trabecular number and pattern factor (P<0.05) in the Enpp1-/- mice in comparison to the wild-type (WT) mice. Mechanical testing revealed that in the WT mice, treatment with etidronate significantly improved work to fracture and increased work post-failure (P<0.05, in comparison to the vehicle-treated WT mice). This significant increase, however, was not observed in the Enpp1-/- mice. Treatment with etidronate had no effect on bone parameters in the WT mice; however, the Enpp1-/- mice displayed an increased structural model index (SMI; P<0.05). We used a recently developed 3D µCT protocol to reconstruct and quantify the extensive aortic calcification in Enpp1-/- mice in comparison to the WT mice. However, treatment with etidronate did not prevent de novo calcification, and did not arrest the progression of established calcification of the aorta.
Collapse
Affiliation(s)
- Carmen Huesa
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| | | | - Jose Luis Millán
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Vicky E MacRae
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
175
|
Counter-regulatory phosphatases TNAP and NPP1 temporally regulate tooth root cementogenesis. Int J Oral Sci 2015; 7:27-41. [PMID: 25504209 PMCID: PMC4817535 DOI: 10.1038/ijos.2014.62] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2014] [Indexed: 12/29/2022] Open
Abstract
Cementum is critical for anchoring the insertion of periodontal ligament fibers to the tooth root. Several aspects of cementogenesis remain unclear, including differences between acellular cementum and cellular cementum, and between cementum and bone. Biomineralization is regulated by the ratio of inorganic phosphate (Pi) to mineral inhibitor pyrophosphate (PPi), where local Pi and PPi concentrations are controlled by phosphatases including tissue-nonspecific alkaline phosphatase (TNAP) and ectonucleotide pyrophosphatase/phosphodiesterase 1 (NPP1). The focus of this study was to define the roles of these phosphatases in cementogenesis. TNAP was associated with earliest cementoblasts near forming acellular and cellular cementum. With loss of TNAP in the Alpl null mouse, acellular cementum was inhibited, while cellular cementum production increased, albeit as hypomineralized cementoid. In contrast, NPP1 was detected in cementoblasts after acellular cementum formation, and at low levels around cellular cementum. Loss of NPP1 in the Enpp1 null mouse increased acellular cementum, with little effect on cellular cementum. Developmental patterns were recapitulated in a mouse model for acellular cementum regeneration, with early TNAP expression and later NPP1 expression. In vitro, cementoblasts expressed Alpl gene/protein early, whereas Enpp1 gene/protein expression was significantly induced only under mineralization conditions. These patterns were confirmed in human teeth, including widespread TNAP, and NPP1 restricted to cementoblasts lining acellular cementum. These studies suggest that early TNAP expression creates a low PPi environment promoting acellular cementum initiation, while later NPP1 expression increases PPi, restricting acellular cementum apposition. Alterations in PPi have little effect on cellular cementum formation, though matrix mineralization is affected.
Collapse
|
176
|
Foster BL, Sheen CR, Hatch NE, Liu J, Cory E, Narisawa S, Kiffer-Moreira T, Sah RL, Whyte MP, Somerman MJ, Millán JL. Periodontal Defects in the A116T Knock-in Murine Model of Odontohypophosphatasia. J Dent Res 2015; 94:706-14. [PMID: 25716980 DOI: 10.1177/0022034515573273] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mutations in ALPL result in hypophosphatasia (HPP), a disease causing defective skeletal mineralization. ALPL encodes tissue nonspecific alkaline phosphatase (ALP), an enzyme that promotes mineralization by reducing inorganic pyrophosphate, a mineralization inhibitor. In addition to skeletal defects, HPP causes dental defects, and a mild clinical form of HPP, odontohypophosphatasia, features only a dental phenotype. The Alpl knockout (Alpl (-/-)) mouse phenocopies severe infantile HPP, including profound skeletal and dental defects. However, the severity of disease in Alpl (-/-) mice prevents analysis at advanced ages, including studies to target rescue of dental tissues. We aimed to generate a knock-in mouse model of odontohypophosphatasia with a primarily dental phenotype, based on a mutation (c.346G>A) identified in a human kindred with autosomal dominant odontohypophosphatasia. Biochemical, skeletal, and dental analyses were performed on the resulting Alpl(+/A116T) mice to validate this model. Alpl(+/A116T) mice featured 50% reduction in plasma ALP activity compared with wild-type controls. No differences in litter size, survival, or body weight were observed in Alpl(+/A116T) versus wild-type mice. The postcranial skeleton of Alpl(+/A116T) mice was normal by radiography, with no differences in femur length, cortical/trabecular structure or mineral density, or mechanical properties. Parietal bone trabecular compartment was mildly altered. Alpl(+/A116T) mice featured alterations in the alveolar bone, including radiolucencies and resorptive lesions, osteoid accumulation on the alveolar bone crest, and significant differences in several bone properties measured by micro-computed tomography. Nonsignificant changes in acellular cementum did not appear to affect periodontal attachment or function, although circulating ALP activity was correlated significantly with incisor cementum thickness. The Alpl(+/A116T) mouse is the first model of odontohypophosphatasia, providing insights on dentoalveolar development and function under reduced ALP, bringing attention to direct effects of HPP on alveolar bone, and offering a new model for testing potential dental-targeted therapies in future studies.
Collapse
Affiliation(s)
- B L Foster
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - C R Sheen
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - N E Hatch
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - J Liu
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - E Cory
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - S Narisawa
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - T Kiffer-Moreira
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - R L Sah
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - M P Whyte
- Center for Metabolic Bone Disease and Molecular Research, Shriners Hospital for Children, St. Louis, MO, USA Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO, USA
| | - M J Somerman
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - J L Millán
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| |
Collapse
|
177
|
Evrard S, Delanaye P, Kamel S, Cristol JP, Cavalier E. Vascular calcification: from pathophysiology to biomarkers. Clin Chim Acta 2015; 438:401-14. [PMID: 25236333 DOI: 10.1016/j.cca.2014.08.034] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 08/27/2014] [Accepted: 08/29/2014] [Indexed: 02/06/2023]
Abstract
The link between vascular calcification (VC) and increased mortality is now well established. Over time, as clinical importance of this phenomenon has begun to be fully considered, scientists have highlighted more and more physiopathological mechanisms and signaling pathways that underlie VC. Several conditions such as diabetes, dyslipidemia and renal diseases are undoubtedly identified as predisposing factors. But even if the process is better understood, many questions still remain unanswered. This review briefly develops the various theories that attempt to explain mineralization genesis. Nonetheless, the main purpose of the article is to provide a profile of the various existing biomarkers of VC. Indeed, in the past years, a lot of inhibitors and promoters, which form a dense and interconnected network, were identified. Given importance to assess and control mineralization process, a focusing on accumulated knowledge of each marker seemed to be necessary. Therefore, we tried to define their respective role in the physiopathology and how they can contribute to calcification risk assessment. Among these, Klotho/fibroblast growth factor-23, fetuin-A, Matrix Gla protein, Bone morphogenetic protein-2, osteoprotegerin, osteopontin, osteonectin, osteocalcin, pyrophosphate and sclerostin are specifically discussed.
Collapse
Affiliation(s)
- Séverine Evrard
- Department of Clinical Chemistry, University of Liège, CHU Sart-Tilman, Liège, Belgium
| | - Pierre Delanaye
- Department of Nephrology, Dialysis and Hypertension, University of Liège, CHU Sart-Tilman, Liège, Belgium
| | - Said Kamel
- Laboratoire de Biochimie, CHU Amiens, Amiens, France; INSERM U1088, Université de Picardie Jules-Verne, Amiens, France
| | - Jean-Paul Cristol
- Laboratoire de Biochimie, CHRU de Montpellier, Hôpital Lapeyronie, Montpellier, France
| | - Etienne Cavalier
- Department of Clinical Chemistry, University of Liège, CHU Sart-Tilman, Liège, Belgium.
| |
Collapse
|
178
|
|
179
|
Abstract
Genetically modified mice are powerful tools for understanding the functions of genes and proteins and often serve as models of human disease. Here, several knockout and transgenic mouse lines related to tissue-nonspecific alkaline phosphatase (TNAP) are described. Conventional TNAP knockout mice die before weaning and show vitamin B6 dependent epilepsy and impaired bone mineralization, mimicking infantile hypophosphatasia. Administration of recombinant human TNAP rescues the lethal phenotype and improves bone mineralization in the null knockout mice, and this enzyme replacement therapy has been successfully applied to the treatment of human patients. Transgenic expression of human TNAP also rescues the TNAP knockout mice. Studies of the TNAP knockout mice and their double knockouts with ectonucleotide pyrophosphatase/phosphodiesterase 1 or progressive ankylosis protein revealed that pyridoxal phosphate and inorganic pyrophosphate are natural substrates of TNAP. Bone osteopontin from TNAP knockout mice is highly phosphorylated, whereas osteopontin from TNAP knockout mice expressing human TNAP is de-phosphorylated, similar to that in wild type mice, indicating that osteopontin is also a natural substrate of TNAP and that phosphorylated osteopontin contributes the impaired bone mineralization in TNAP knockout mice. Conditional TNAP knockout mice and TNAP mutants produced by ENU (N-ethyl-N-nitrosourea) mutagenesis show milder hypophosphatasia and are expected to be useful models of adult hypophosphatasia.
Collapse
|
180
|
Probenecid as a sensitizer of bisphosphonate-mediated effects in breast cancer cells. Mol Cancer 2014; 13:265. [PMID: 25496233 PMCID: PMC4295226 DOI: 10.1186/1476-4598-13-265] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 11/27/2014] [Indexed: 12/17/2022] Open
Abstract
Background Anti-resorptive bisphosphonates (BP) are used for the treatment of osteoporosis and bone metastases. Clinical studies indicated a benefit in survival and tumor relapse in subpopulations of breast cancer patients receiving zoledronic acid, thus stimulating the debate about its anti-tumor activity. Amino-bisphosphonates in nM concentrations inhibit farnesyl pyrophosphate synthase leading to accumulation of isopentenyl pyrophosphate (IPP) and the ATP/pyrophosphate adduct ApppI, which induces apoptosis in osteoclasts. For anti-tumor effects μM concentrations are needed and a sensitizer for bisphosphonate effects would be beneficial in clinical anti-tumor applications. We hypothesized that enhancing intracellular pyrophosphate accumulation via inhibition of probenecid-sensitive channels and transporters would sensitize tumor cells for bisphosphonates anti-tumor efficacy. Method MDA-MB-231, T47D and MCF-7 breast cancer cells were treated with BP (zoledronic acid, risedronate, ibandronate, alendronate) and the pyrophosphate channel inhibitors probenecid and novobiocin. We determined cell viability and caspase 3/7 activity (apoptosis), accumulation of IPP and ApppI, expression of ANKH, PANX1, ABCC1, SLC22A11, and the zoledronic acid target gene and tumor-suppressor KLF2. Results Treatment of MDA-MB-231 with BP induced caspase 3/7 activity, with zoledronic acid being the most effective. In MCF-7 and T47D either BP markedly suppressed cell viability with only minor effects on apoptosis. Co-treatment with probenecid enhanced BP effects on cell viability, IPP/ApppI accumulation as measurable in MCF-7 and T47D cells, caspase 3/7 activity and target gene expression. Novobiocin co-treatment of MDA-MB-231 yielded identical results on viability and apoptosis compared to probenecid, rendering SLC22A family members as candidate modulators of BP effects, whereas no such evidence was found for ANKH, ABCC1 and PANX1. Conclusions In summary, we demonstrate effects of various bisphosphonates on caspase 3/7 activity, cell viability and expression of tumor suppressor genes in breast cancer cells. Blocking probenecid and novobiocin-sensitive channels and transporters enhances BP anti-tumor effects and renders SLC22A family members as good candidates as BP modulators. Further studies will have to unravel if treatment with such BP-sensitizers translates into preclinical and clinical efficacy. Electronic supplementary material The online version of this article (doi:10.1186/1476-4598-13-265) contains supplementary material, which is available to authorized users.
Collapse
|
181
|
Shimokado A, Sun Y, Nakanishi M, Sato F, Oikawa K, Akasaka T, Muragaki Y. Smad3 plays an inhibitory role in phosphate-induced vascular smooth muscle cell calcification. Exp Mol Pathol 2014; 97:458-64. [DOI: 10.1016/j.yexmp.2014.10.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 09/10/2014] [Accepted: 10/06/2014] [Indexed: 01/10/2023]
|
182
|
Yadav MC, Huesa C, Narisawa S, Hoylaerts MF, Moreau A, Farquharson C, Millán JL. Ablation of osteopontin improves the skeletal phenotype of phospho1(-/-) mice. J Bone Miner Res 2014; 29:2369-81. [PMID: 24825455 PMCID: PMC5247257 DOI: 10.1002/jbmr.2281] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Revised: 05/01/2014] [Accepted: 05/07/2014] [Indexed: 11/06/2022]
Abstract
PHOSPHO1 and tissue-nonspecific alkaline phosphatase (TNAP) have nonredundant functions during skeletal mineralization. Although TNAP deficiency (Alpl(-/-) mice) leads to hypophosphatasia, caused by accumulation of the mineralization inhibitor inorganic pyrophosphate (PPi ), comparably elevated levels of PPi in Phospho1(-/-) mice do not explain their stunted growth, spontaneous fractures, bowed long bones, osteomalacia, and scoliosis. We have previously shown that elevated PPi in Alpl(-/-) mice is accompanied by elevated osteopontin (OPN), another potent mineralization inhibitor, and that the amount of OPN correlates with the severity of hypophosphatasia in mice. Here we demonstrate that plasma OPN is elevated and OPN expression is upregulated in the skeleton, particularly in the vertebrae, of Phospho1(-/-) mice. Liquid chromatography/tandem mass spectrometry showed an increased proportion of phosphorylated OPN (p-OPN) peptides in Phospho1(-/-) mice, suggesting that accumulation of p-OPN causes the skeletal abnormalities in Phospho1(-/-) mice. We also show that ablation of the OPN gene, Spp1, leads to improvements in the skeletal phenotype in Phospho1(-/-) as they age. In particular, their scoliosis is ameliorated at 1 month of age and is completely rescued at 3 months of age. There is also improvement in the long bone defects characteristic of Phospho1(-/-) mice at 3 months of age. Mineralization assays comparing [Phospho1(-/-) ; Spp1(-/-) ], Phospho1(-/-) , and Spp1(-/-) chondrocytes display corrected mineralization by the double knockout cells. Expression of chondrocyte differentiation markers was also normalized in the [Phospho1(-/-) ; Spp1(-/-) ] mice. Thus, although Alpl and Phospho1 deficiencies lead to similar skeletal phenotypes and comparable changes in the expression levels of PPi and OPN, there is a clear dissociation in the hierarchical roles of these potent inhibitors of mineralization, with elevated PPi and elevated p-OPN levels causing the respective skeletal phenotypes in Alpl(-/-) and Phospho1(-/-) mice.
Collapse
Affiliation(s)
- Manisha C Yadav
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | | | | | | | | | | | | |
Collapse
|
183
|
Huesa C, Zhu D, Glover JD, Ferron M, Karsenty G, Milne EM, Millan JL, Ahmed SF, Farquharson C, Morton NM, MacRae VE. Deficiency of the bone mineralization inhibitor NPP1 protects mice against obesity and diabetes. Dis Model Mech 2014; 7:1341-50. [PMID: 25368121 PMCID: PMC4257003 DOI: 10.1242/dmm.017905] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The emergence of bone as an endocrine regulator has prompted a re-evaluation of the role of bone mineralization factors in the development of metabolic disease. Ectonucleotide pyrophosphatase/phosphodiesterase-1 (NPP1) controls bone mineralization through the generation of pyrophosphate, and levels of NPP1 are elevated both in dermal fibroblast cultures and muscle of individuals with insulin resistance. We investigated the metabolic phenotype associated with impaired bone metabolism in mice lacking the gene that encodes NPP1 (Enpp1−/− mice). Enpp1−/− mice exhibited mildly improved glucose homeostasis on a normal diet but showed a pronounced resistance to obesity and insulin resistance in response to chronic high-fat feeding. Enpp1−/− mice had increased levels of the insulin-sensitizing bone-derived hormone osteocalcin but unchanged insulin signalling within osteoblasts. A fuller understanding of the pathways of NPP1 could inform the development of novel therapeutic strategies for treating insulin resistance.
Collapse
Affiliation(s)
- Carmen Huesa
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh, EH25 9RG, UK
| | - Dongxing Zhu
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh, EH25 9RG, UK.
| | - James D Glover
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh, EH25 9RG, UK
| | - Mathieu Ferron
- Integrative and Molecular Physiology Research Unit Institut de Recherches Cliniques de Montréal (IRCM), 110 Avenue des Pins Ouest - Laboratory 2750, Montréal, QC H2W 1R7, Canada
| | - Gerard Karsenty
- Department of Developmental Genetics, Columbia University, NY 10032, USA
| | - Elspeth M Milne
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh, EH25 9RG, UK
| | - José Luis Millan
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - S Faisal Ahmed
- Developmental Medicine, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Colin Farquharson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh, EH25 9RG, UK
| | - Nicholas M Morton
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Vicky E MacRae
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh, EH25 9RG, UK
| |
Collapse
|
184
|
Molecular, phenotypic aspects and therapeutic horizons of rare genetic bone disorders. BIOMED RESEARCH INTERNATIONAL 2014; 2014:670842. [PMID: 25530967 PMCID: PMC4230237 DOI: 10.1155/2014/670842] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 08/12/2014] [Accepted: 08/24/2014] [Indexed: 12/21/2022]
Abstract
A rare disease afflicts less than 200,000 individuals, according to the National Organization for Rare Diseases (NORD) of the United States. Over 6,000 rare disorders affect approximately 1 in 10 Americans. Rare genetic bone disorders remain the major causes of disability in US patients. These rare bone disorders also represent a therapeutic challenge for clinicians, due to lack of understanding of underlying mechanisms. This systematic review explored current literature on therapeutic directions for the following rare genetic bone disorders: fibrous dysplasia, Gorham-Stout syndrome, fibrodysplasia ossificans progressiva, melorheostosis, multiple hereditary exostosis, osteogenesis imperfecta, craniometaphyseal dysplasia, achondroplasia, and hypophosphatasia. The disease mechanisms of Gorham-Stout disease, melorheostosis, and multiple hereditary exostosis are not fully elucidated. Inhibitors of the ACVR1/ALK2 pathway may serve as possible therapeutic intervention for FOP. The use of bisphosphonates and IL-6 inhibitors has been explored to be useful in the treatment of fibrous dysplasia, but more research is warranted. Cell therapy, bisphosphonate polytherapy, and human growth hormone may avert the pathology in osteogenesis imperfecta, but further studies are needed. There are still no current effective treatments for these bone disorders; however, significant promising advances in therapeutic modalities were developed that will limit patient suffering and treat their skeletal disabilities.
Collapse
|
185
|
Liu J, Nam HK, Campbell C, Gasque KCDS, Millán JL, Hatch NE. Tissue-nonspecific alkaline phosphatase deficiency causes abnormal craniofacial bone development in the Alpl(-/-) mouse model of infantile hypophosphatasia. Bone 2014; 67:81-94. [PMID: 25014884 PMCID: PMC4149826 DOI: 10.1016/j.bone.2014.06.040] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 06/09/2014] [Accepted: 06/30/2014] [Indexed: 12/27/2022]
Abstract
UNLABELLED Tissue-nonspecific alkaline phosphatase (TNAP) is an enzyme present on the surface of mineralizing cells and their derived matrix vesicles that promotes hydroxyapatite crystal growth. Hypophosphatasia (HPP) is an inborn-error-of-metabolism that, dependent upon age of onset, features rickets or osteomalacia due to loss-of function mutations in the gene (Alpl) encoding TNAP. Craniosynostosis is prevalent in infants with HPP and other forms of rachitic disease but how craniosynostosis develops in these disorders is unknown. OBJECTIVES Because craniosynostosis carries high morbidity, we are investigating craniofacial skeletal abnormalities in Alpl(-/-) mice to establish these mice as a model of HPP-associated craniosynostosis and determine mechanisms by which TNAP influences craniofacial skeletal development. METHODS Cranial bone, cranial suture and cranial base abnormalities were analyzed by micro-CT and histology. Craniofacial shape abnormalities were quantified using digital calipers. TNAP expression was suppressed in MC3T3E1(C4) calvarial cells by TNAP-specific shRNA. Cells were analyzed for changes in mineralization, gene expression, proliferation, apoptosis, matrix deposition and cell adhesion. RESULTS Alpl(-/-) mice feature craniofacial shape abnormalities suggestive of limited anterior-posterior growth. Craniosynostosis in the form of bony coronal suture fusion is present by three weeks after birth. Alpl(-/-) mice also exhibit marked histologic abnormalities of calvarial bones and the cranial base involving growth plates, cortical and trabecular bone within two weeks of birth. Analysis of calvarial cells in which TNAP expression was suppressed by shRNA indicates that TNAP deficiency promotes aberrant osteoblastic gene expression, diminished matrix deposition, diminished proliferation, increased apoptosis and increased cell adhesion. CONCLUSIONS These findings demonstrate that Alpl(-/-) mice exhibit a craniofacial skeletal phenotype similar to that seen in infants with HPP, including true bony craniosynostosis in the context of severely diminished bone mineralization. Future studies will be required to determine if TNAP deficiency and other forms of rickets promote craniosynostosis directly through abnormal calvarial cell behavior, or indirectly due to deficient growth of the cranial base.
Collapse
Affiliation(s)
- Jin Liu
- 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
| | - Cassie Campbell
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Nan E Hatch
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
186
|
Dabisch-Ruthe M, Brock A, Kuzaj P, Charbel Issa P, Szliska C, Knabbe C, Hendig D. Variants in genes encoding pyrophosphate metabolizing enzymes are associated with Pseudoxanthoma elasticum. Clin Biochem 2014; 47:60-7. [DOI: 10.1016/j.clinbiochem.2014.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 06/30/2014] [Accepted: 07/03/2014] [Indexed: 10/25/2022]
|
187
|
Sliwiński L, Folwarczna J, Pytlik M, Cegieła U, Nowińska B, Trzeciak H, Trzeciak HI. Do effects of propranolol on the skeletal system depend on the estrogen status? Pharmacol Rep 2014; 65:1345-56. [PMID: 24399731 DOI: 10.1016/s1734-1140(13)71493-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 06/11/2013] [Indexed: 11/25/2022]
Abstract
BACKGROUND Propranolol, a nonselective β-adrenergic receptor antagonist, was reported to favorably affect the skeletal system in different animal models. The aim of the study was to investigate whether the effects of propranolol on the skeletal system depend on the estrogen status. METHODS The in vivo experiments were carried out on the following groups of mature female Wistar rats: sham-operated control rats, sham-operated rats receiving propranolol, ovariectomized (OVX) control rats, OVX rats receiving propranolol, OVX rats receiving estradiol, OVX rats receiving estradiol and propranolol. Propranolol hydrochloride (10 mg/kg po) and/or estradiol (0.1 mg/kg po) were administered daily for 4 weeks. Bone mass, mineral and calcium content, macrometric and histomorphometric parameters, and mechanical properties were examined. In vitro, effects of estradiol and propranolol on the formation of mouse osteoclasts and on the mRNA expression of genes related to osteoclastogenesis, bone formation and mineralization, as well as adrenergic and estrogen signalling in mouse osteoblasts were investigated. RESULTS AND CONCLUSION Propranolol exerted some favorable effects on the rat skeletal system in vivo, independently of the estrogen status. However, in vitro studies indicated a possibility of some antagonistic relations between the estradiol and propranolol effects.
Collapse
Affiliation(s)
- Leszek Sliwiński
- Department of Pharmacology, Medical University of Silesia, Katowice, Jagiellońska 4, PL 41-200 Sosnowiec, Poland.
| | | | | | | | | | | | | |
Collapse
|
188
|
Kuzynski M, Goss M, Bottini M, Yadav MC, Mobley C, Winters T, Poliard A, Kellermann O, Lee B, Millan JL, Napierala D. Dual role of the Trps1 transcription factor in dentin mineralization. J Biol Chem 2014; 289:27481-93. [PMID: 25128529 DOI: 10.1074/jbc.m114.550129] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
TRPS1 (tricho-rhino-phalangeal syndrome) is a unique GATA-type transcription factor that acts as a transcriptional repressor. TRPS1 deficiency and dysregulated TRPS1 expression result in skeletal and dental abnormalities implicating TRPS1 in endochondral bone formation and tooth development. Moreover, patients with tricho-rhino-phalangeal syndrome frequently present with low bone mass indicating TRPS1 involvement in bone homeostasis. In addition, our previous data demonstrated accelerated mineralization of the perichondrium in Trps1 mutant mice and impaired dentin mineralization in Col1a1-Trps1 transgenic mice, implicating Trps1 in the mineralization process. To understand the role of Trps1 in the differentiation and function of cells producing mineralized matrix, we used a preodontoblastic cell line as a model of dentin mineralization. We generated both Trps1-deficient and Trps1-overexpressing stable cell lines and analyzed the progression of mineralization by alkaline phosphatase and alizarin red staining. As predicted, based on our previous in vivo data, delayed and decreased mineralization of Trps1-overexpressing odontoblastic cells was observed when compared with control cells. This was associated with down-regulation of genes regulating phosphate homeostasis. Interestingly, Trps1-deficient cells lost the ability to mineralize and demonstrated decreased expression of several genes critical for initiating the mineralization process, including Alpl and Phospho1. Based on these data, we have concluded that Trps1 serves two critical and context-dependent functions in odontoblast-regulated mineralization as follows: 1) Trps1 is required for odontoblast maturation by supporting expression of genes crucial for initiating the mineralization process, and 2) Trps1 represses the function of mature cells and, consequently, restricts the extent of extracellular matrix mineralization.
Collapse
Affiliation(s)
- Maria Kuzynski
- From the Institute of Oral Health Research, Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Morgan Goss
- From the Institute of Oral Health Research, Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Massimo Bottini
- the Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037, the Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133Rome, Italy
| | - Manisha C Yadav
- the Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Callie Mobley
- From the Institute of Oral Health Research, Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Tony Winters
- From the Institute of Oral Health Research, Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Anne Poliard
- the EA2496 UFR d'Odontologie, Université Paris Descartes, 92120 Montrouge, France
| | - Odile Kellermann
- INSERM UMR-S 1124, Université René Descartes Paris 5, Centre Universitaire des Saints-Pères, 75270 Paris Cedex 06, France
| | - Brendan Lee
- the Department of Molecular and Human Genetics, Baylor College of Medicine, and the Howard Hughes Medical Institute, Houston, Texas 77030
| | - Jose Luis Millan
- the Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037
| | - Dobrawa Napierala
- From the Institute of Oral Health Research, Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Alabama at Birmingham, Birmingham, Alabama 35294,
| |
Collapse
|
189
|
de la Croix Ndong J, Makowski AJ, Uppuganti S, Vignaux G, Ono K, Perrien DS, Joubert S, Baglio SR, Granchi D, Stevenson DA, Rios JJ, Nyman JS, Elefteriou F. Asfotase-α improves bone growth, mineralization and strength in mouse models of neurofibromatosis type-1. Nat Med 2014; 20:904-10. [PMID: 24997609 PMCID: PMC4126855 DOI: 10.1038/nm.3583] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 05/01/2014] [Indexed: 12/20/2022]
Abstract
Mineralization of the skeleton depends on the balance between levels of
pyrophosphate (PPi), an inhibitor of hydroxyapatite formation, and phosphate generated
from PPi breakdown by alkaline phosphatase (ALP). We report here that ablation of
Nf1, encoding the RAS/GTPase–activating protein neurofibromin,
in bone–forming cells leads to supraphysiologic PPi accumulation, caused by a
chronic ERK–dependent increase in genes promoting PPi synthesis and extracellular
transport, namely Enpp1 and Ank. It also prevents
BMP2–induced osteoprogenitor differentiation and, consequently, expression of ALP
and PPi breakdown, further contributing to PPi accumulation. The short stature, impaired
bone mineralization and strength in mice lacking Nf1 in
osteochondroprogenitors or osteoblasts could be corrected by enzyme therapy aimed at
reducing PPi concentration. These results establish neurofibromin as an essential
regulator of bone mineralization, suggest that altered PPi homeostasis contributes to the
skeletal dysplasiae associated with neurofibromatosis type-1 (NF1), and that some of the
NF1 skeletal conditions might be preventable pharmacologically.
Collapse
Affiliation(s)
- Jean de la Croix Ndong
- 1] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [2] Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Alexander J Makowski
- 1] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [2] Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA. [3] Department of Orthopaedic Surgery &Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [4] Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Sasidhar Uppuganti
- 1] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [2] Department of Orthopaedic Surgery &Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Guillaume Vignaux
- 1] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [2] Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Koichiro Ono
- 1] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [2] Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [3] Department of Orthopaedics, Nohon Koukan Hospital, Kawasaki, Kanagawa, Japan
| | - Daniel S Perrien
- 1] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [2] Department of Orthopaedic Surgery &Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [3] Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA. [4] Vanderbilt University Institute of Imaging Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Serena R Baglio
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Donatella Granchi
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - David A Stevenson
- Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
| | - Jonathan J Rios
- 1] Sarah M. and Charles E. Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, Texas, USA. [2] Department of Pediatrics, UT Southwestern Medical Center, Dallas, Texas, USA. [3] Eugene McDermott Center for Human Growth &Development, UT Southwestern Medical Center, Dallas, Texas, USA. [4] Department of Orthopaedic Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jeffry S Nyman
- 1] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [2] Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA. [3] Department of Orthopaedic Surgery &Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [4] Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Florent Elefteriou
- 1] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [2] Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [3] Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [4] Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
190
|
Rathan S, Yoganathan AP, O’Neill WC. The role of inorganic pyrophosphate in aortic valve calcification. THE JOURNAL OF HEART VALVE DISEASE 2014; 23:387-394. [PMID: 25803964 PMCID: PMC5463180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
BACKGROUND AND AIM OF THE STUDY Aortic valve (AV) calcification is a major cause of morbidity and mortality, yet the molecular mechanisms involved are poorly understood. Hence, an ex vivo model of calcification in intact AVs was developed in order to test the role of orthophosphate and pyrophosphate (PPi), both of which factors are known to influence vascular calcification. METHODS Porcine AV leaflets were cultured in serum-free medium under static conditions for eight days, over which time leaflet architecture and viability were preserved. Calcification was measured as the incorporation of 45Ca, with confirmation by Alizarin Red staining. RESULTS Calcification required both a high phosphate concentration (3.8 mM) and removal of PPi with alkaline phosphatase or inorganic pyrophosphatase. Calcification occurred predominantly on the fibrosa and was arrested by the bisphosphonate etidronate, a non-hydrolyzable analog of PPi. Leaflets released PPi into the medium, and this was enhanced by MLS38949, a specific inhibitor of tissue non-specific alkaline phosphatase (TNAP). Furthermore, leaflets synthesized PPi from extracellular ATP, which was reduced by β,γ-methylene-ATP, an inhibitor of ectonucleotide pyrophosphorylase phosphodiesterase (NPP1). CONCLUSION The ex vivo AV calcification model developed in the present study showed that extracellular PPi, produced by valvular tissue, is a potent inhibitor of valvular calcification. In addition to synthesis, hydrolysis by TNAP also controls PPi levels and calcification. The results suggest that a decreased synthesis or increased hydrolysis of pyrophosphate may contribute to valvular calcification, and that bisphosphonates or inhibitors of TNAP are potential preventive strategies of the process. TNAP are potential preventive strategies.
Collapse
Affiliation(s)
- Swetha Rathan
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA
| | - Ajit P. Yoganathan
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA
| | | |
Collapse
|
191
|
Brie IC, Soritau O, Dirzu N, Berce C, Vulpoi A, Popa C, Todea M, Simon S, Perde-Schrepler M, Virag P, Barbos O, Chereches G, Berce P, Cernea V. Comparative in vitro study regarding the biocompatibility of titanium-base composites infiltrated with hydroxyapatite or silicatitanate. J Biol Eng 2014; 8:14. [PMID: 24987458 PMCID: PMC4077223 DOI: 10.1186/1754-1611-8-14] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 06/13/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The development of novel biomaterials able to control cell activities and direct their fate is warranted for engineering functional bone tissues. Adding bioactive materials can improve new bone formation and better osseointegration. Three types of titanium (Ti) implants were tested for in vitro biocompatibility in this comparative study: Ti6Al7Nb implants with 25% total porosity used as controls, implants infiltrated using a sol-gel method with hydroxyapatite (Ti HA) and silicatitanate (Ti SiO2). The behavior of human osteoblasts was observed in terms of adhesion, cell growth and differentiation. RESULTS The two coating methods have provided different morphological and chemical properties (SEM and EDX analysis). Cell attachment in the first hour was slower on the Ti HA scaffolds when compared to Ti SiO2 and porous uncoated Ti implants. The Alamar blue test and the assessment of total protein content uncovered a peak of metabolic activity at day 8-9 with an advantage for Ti SiO2 implants. Osteoblast differentiation and de novo mineralization, evaluated by osteopontin (OP) expression (ELISA and immnocytochemistry), alkaline phosphatase (ALP) activity, calcium deposition (alizarin red), collagen synthesis (SIRCOL test and immnocytochemical staining) and osteocalcin (OC) expression, highlighted the higher osteoconductive ability of Ti HA implants. Higher soluble collagen levels were found for cells cultured in simple osteogenic differentiation medium on control Ti and Ti SiO2 implants. Osteocalcin (OC), a marker of terminal osteoblastic differentiation, was most strongly expressed in osteoblasts cultivated on Ti SiO2 implants. CONCLUSIONS The behavior of osteoblasts depends on the type of implant and culture conditions. Ti SiO2 scaffolds sustain osteoblast adhesion and promote differentiation with increased collagen and non-collagenic proteins (OP and OC) production. Ti HA implants have a lower ability to induce cell adhesion and proliferation but an increased capacity to induce early mineralization. Addition of growth factors BMP-2 and TGFβ1 in differentiation medium did not improve the mineralization process. Both types of infiltrates have their advantages and limitations, which can be exploited depending on local conditions of bone lesions that have to be repaired. These limitations can also be offset through methods of functionalization with biomolecules involved in osteogenesis.
Collapse
Affiliation(s)
- Ioana-Carmen Brie
- The Institute of Oncology "Prof. Dr. I. Chiricuta" Cluj-Napoca, Cluj-Napoca, Romania ; University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj-Napoca, Cluj-Napoca, Romania
| | - Olga Soritau
- The Institute of Oncology "Prof. Dr. I. Chiricuta" Cluj-Napoca, Cluj-Napoca, Romania
| | | | - Cristian Berce
- University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj-Napoca, Cluj-Napoca, Romania
| | - Adriana Vulpoi
- Faculty of Physics & Institute of Interdisciplinary Research in Bio-Nano-Sciences, Babes Bolyai University, 400084 Cluj-Napoca, Romania
| | | | - Milica Todea
- Faculty of Physics & Institute of Interdisciplinary Research in Bio-Nano-Sciences, Babes Bolyai University, 400084 Cluj-Napoca, Romania
| | - Simion Simon
- Faculty of Physics & Institute of Interdisciplinary Research in Bio-Nano-Sciences, Babes Bolyai University, 400084 Cluj-Napoca, Romania
| | - Maria Perde-Schrepler
- The Institute of Oncology "Prof. Dr. I. Chiricuta" Cluj-Napoca, Cluj-Napoca, Romania
| | - Piroska Virag
- The Institute of Oncology "Prof. Dr. I. Chiricuta" Cluj-Napoca, Cluj-Napoca, Romania
| | - Otilia Barbos
- The Institute of Oncology "Prof. Dr. I. Chiricuta" Cluj-Napoca, Cluj-Napoca, Romania
| | - Gabriela Chereches
- The Institute of Oncology "Prof. Dr. I. Chiricuta" Cluj-Napoca, Cluj-Napoca, Romania
| | | | - Valentin Cernea
- The Institute of Oncology "Prof. Dr. I. Chiricuta" Cluj-Napoca, Cluj-Napoca, Romania ; University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj-Napoca, Cluj-Napoca, Romania
| |
Collapse
|
192
|
Dabisch-Ruthe M, Kuzaj P, Götting C, Knabbe C, Hendig D. Pyrophosphates as a major inhibitor of matrix calcification in Pseudoxanthoma elasticum. J Dermatol Sci 2014; 75:109-20. [PMID: 24907773 DOI: 10.1016/j.jdermsci.2014.04.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 04/22/2014] [Accepted: 04/23/2014] [Indexed: 11/18/2022]
Abstract
BACKGROUND Pseudoxanthoma elasticum (PXE) is a rare hereditary disorder characterized by late onset and progressive calcification of elastic fibers in skin, eyes and the cardiovascular system, exemplifying a model for conditions characterized by soft tissue calcification. OBJECTIVE The aim of our study was to characterize cellular inorganic pyrophosphate (PPi) homeostasis in PXE. METHODS Gene expression of PPi metabolizing enzymes was determined by quantitative real-time PCR after incubation up to 21 days with or without addition of Na2HPO4. Extracellular and cytosolic PPi concentrations were measured by enzyme-linked bioluminescence assay. ALP and ENPP1 activity was determined spectrophotometrically. We further established a human cell culture model suitable for investigating PXE and related disorders without addition of artificial calcification triggers. RESULTS Independently of the experimental conditions, PXE fibroblasts revealed a higher degree of matrix calcification. We observed that matrix calcification was associated with altered gene expression of PPi metabolizing enzymes in PXE fibroblasts. In this context, PXE fibroblasts exhibited significantly higher expression of ALP and OPN and reduced mRNA expression and activity of ENPP1. Here, for the first time cytosolic and extracellular PPi levels were shown to be strongly reduced in PXE fibroblasts. We further showed that PPi concentration in bovine and human sera additives had a strong impact on matrix calcification. In a last experimental line, we demonstrated that addition of PPi analogs reduced matrix calcification of PXE fibroblasts most likely by reducing ALP and OPN mRNA expression, restoring ENPP1 activity and subsequently elevating PPi concentrations. CONCLUSION The results of our study along with recent findings point to the essential role of PPi as the central regulatory metabolites preventing matrix calcification in PXE. But what remains to be determined is the underlying molecular mechanism leading to depletion of PPi in PXE. We further suggest that supplementation of PPi analogs might counteract pathological calcification in PXE and related disorders.
Collapse
Affiliation(s)
- Mareike Dabisch-Ruthe
- Institut für Laboratoriums- und Transfusionsmedizin, Herz- und Diabeteszentrum Nordrhein-Westfalen, Universitätsklinik der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Patricia Kuzaj
- Institut für Laboratoriums- und Transfusionsmedizin, Herz- und Diabeteszentrum Nordrhein-Westfalen, Universitätsklinik der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | | | - Cornelius Knabbe
- Institut für Laboratoriums- und Transfusionsmedizin, Herz- und Diabeteszentrum Nordrhein-Westfalen, Universitätsklinik der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Doris Hendig
- Institut für Laboratoriums- und Transfusionsmedizin, Herz- und Diabeteszentrum Nordrhein-Westfalen, Universitätsklinik der Ruhr-Universität Bochum, Bad Oeynhausen, Germany.
| |
Collapse
|
193
|
Rendenbach C, Yorgan TA, Heckt T, Otto B, Baldauf C, Jeschke A, Streichert T, David JP, Amling M, Schinke T. Effects of extracellular phosphate on gene expression in murine osteoblasts. Calcif Tissue Int 2014; 94:474-83. [PMID: 24366459 DOI: 10.1007/s00223-013-9831-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 12/01/2013] [Indexed: 02/07/2023]
Abstract
That phosphate homeostasis is tightly linked to skeletal mineralization is probably best underscored by the fact that the phosphaturic hormone FGF23 is primarily expressed by terminally differentiated osteoblasts/osteocytes and that increased circulating FGF23 levels are causative for different types of hypophosphatemic rickets. In contrast, FGF23 inactivation results in hyperphosphatemia, and unexpectedly this phenotype is associated with severe osteomalacia in Fgf23-deficient mice. In this context it is interesting that different cell types have been shown to respond to extracellular phosphate, thereby raising the concept that phosphate can act as a signaling molecule. To identify phosphate-responsive genes in primary murine osteoblasts we performed genome wide expression analysis with cells maintained in medium containing either 1 or 4 mM sodium phosphate for 6 h. As confirmed by qRT-PCR, this analysis revealed that several known osteoblast differentiation markers (Bglap, Ibsp, and Phex) were unaffected by raising extracellular phosphate levels. In contrast, we found that the expression of Enpp1 and Ank, two genes encoding inhibitors of matrix mineralization, was induced by extracellular phosphate, while the expression of Sost and Dkk1, two genes encoding inhibitors of bone formation, was negatively regulated. The ability of osteoblasts to respond to extracellular phosphate was dependent on their differentiation state, and shRNA-dependent repression of the phosphate transporter Slc20a1 in MC3T3-E1 cells partially abolished their molecular response to phosphate. Taken together, our results provide further evidence for a role of extracellular phosphate as a signaling molecule and raise the possibility that severe hyperphosphatemia can negatively affect skeletal mineralization.
Collapse
Affiliation(s)
- C Rendenbach
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, 20246, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Vascular calcification is dependent on plasma levels of pyrophosphate. Kidney Int 2014; 85:1351-6. [PMID: 24717293 PMCID: PMC4308968 DOI: 10.1038/ki.2013.521] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 10/22/2013] [Accepted: 10/31/2013] [Indexed: 11/30/2022]
Abstract
Plasma levels of pyrophosphate, an endogenous inhibitor of vascular calcification, are reduced in end-stage renal disease and correlate inversely with arterial calcification. However, it is not known whether the low plasma levels are directly pathogenic or are merely a marker of reduced tissue levels. This was tested in an animal model in which aortas were transplanted between normal mice and Enpp1−/− mice lacking ectonucleotide pyrophosphatase phosphodiesterase, the enzyme that releases extracellular pyrophosphate. Enpp1−/− mice had very low plasma pyrophosphate and developed aortic calcification by 2 months that was greatly accelerated with a high-phosphate diet. Aortas of Enpp1−/− mice showed no further calcification after transplantation into wild type mice fed a high phosphate diet. Aorta allografts of wild type mice calcified in Enpp1−/− mice but less so than the adjacent recipient Enpp1−/− aorta. Donor and recipient aortic calcium contents did not differ in transplants between wild type and Enpp1−/− mice, demonstrating that transplantation per se did not affect calcification. Histology revealed medial calcification with no signs of rejection. Thus, normal levels of extracellular pyrophosphate are sufficient to prevent vascular calcification and systemic Enpp1 deficiency is sufficient to produce vascular calcification despite normal vascular extracellular pyrophosphate production. This establishes an important role for circulating extracellular pyrophosphate in preventing vascular calcification.
Collapse
|
195
|
Boraldi F, Annovi G, Bartolomeo A, Quaglino D. Fibroblasts from patients affected by Pseudoxanthoma elasticum exhibit an altered PPi metabolism and are more responsive to pro-calcifying stimuli. J Dermatol Sci 2014; 74:72-80. [PMID: 24461675 DOI: 10.1016/j.jdermsci.2013.12.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/13/2013] [Accepted: 12/19/2013] [Indexed: 01/31/2023]
Abstract
BACKGROUND Pseudoxanthoma elasticum (PXE) is a genetic disorder characterized by progressive calcification of soft connective tissues. The pathogenesis is still hard to pin down. In PXE dermal fibroblasts, in addition to impaired carboxylation of the vitamin K-dependent inhibitor matrix Gla protein (MGP), we have also demonstrated an up-regulation of alkaline phosphatase activity. In the light of these data we have suggested that both calcium and phosphate metabolism might be locally altered, both pathways acting in synergy on the occurrence of matrix calcification. OBJECTIVE This study aims to better explore if cultured PXE fibroblasts, compared to control cells, exhibit a modified inorganic pyrophosphate (PPi) metabolism and are more responsive to pro-calcifying stimuli. METHODS Primary human dermal fibroblasts isolated from healthy individuals and from PXE patients were cultured for different time points in standard and in pro-calcifying media. The expression of ANKH/ANKH, ENPP1/PC1, ALPL/TNAP, SPP1/OPN was evaluated by qRT-PCR and Western blot, respectively. TNAP activity was measured by spectrophotometric analyses, whereas calcification was investigated by light and electron microscopy as well as by micro-analytical techniques. RESULTS In the presence of pro-calcifying stimuli, dermal fibroblasts alter their phenotype favouring matrix mineralization. In particular, ENPP1/PC1 and SPP1/OPN expression, as well as TNAP activity, was differently expressed in control and in PXE fibroblasts. Moreover, in pathologic cells the ratio between factors favouring and reducing PPi availability exhibits a more pronounced shift towards a pro-calcifying balance. CONCLUSION PXE fibroblasts are more susceptible to pro-calcifying stimuli and in these cells an altered PPi metabolism contributes to matrix calcification.
Collapse
Affiliation(s)
- Federica Boraldi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Annovi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Angelica Bartolomeo
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Daniela Quaglino
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
196
|
Abstract
Human and mouse alkaline phosphatases (AP) are encoded by a multigene family expressed ubiquitously in multiple tissues. Gene knockout (KO) findings have helped define some of the precise exocytic functions of individual isozymes in bone, teeth, the central nervous system, and in the gut. For instance, deficiency in tissue-nonspecific alkaline phosphatase (TNAP) in mice (Alpl (-/-) mice) and humans leads to hypophosphatasia (HPP), an inborn error of metabolism characterized by epileptic seizures in the most severe cases, caused by abnormal metabolism of pyridoxal-5'-phosphate (the predominant form of vitamin B6) and by hypomineralization of the skeleton and teeth featuring rickets and early loss of teeth in children or osteomalacia and dental problems in adults caused by accumulation of inorganic pyrophosphate (PPi). Enzyme replacement therapy with mineral-targeting TNAP prevented all the manifestations of HPP in mice, and clinical trials with this protein therapeutic are showing promising results in rescuing life-threatening HPP in infants. Conversely, TNAP induction in the vasculature during generalized arterial calcification of infancy (GACI), type II diabetes, obesity, and aging can cause medial vascular calcification. TNAP inhibitors, discussed extensively in this book, are in development to prevent pathological arterial calcification. The brush border enzyme intestinal alkaline phosphatase (IAP) plays an important role in fatty acid (FA) absorption, in protecting gut barrier function, and in determining the composition of the gut microbiota via its ability to dephosphorylate lipopolysaccharide (LPS). Knockout mice (Akp3 (-/-)) deficient in duodenal-specific IAP (dIAP) become obese, and develop hyperlipidemia and hepatic steatosis when fed a high-fat diet (HFD). These changes are accompanied by upregulation in the jejunal-ileal expression of the Akp6 IAP isozyme (global IAP, or gIAP) and concomitant upregulation of FAT/CD36, a phosphorylated fatty acid translocase thought to play a role in facilitating the transport of long-chain fatty acids into cells. gIAP, but not dIAP, is able to modulate the phosphorylation status of FAT/CD36. dIAP, even though it is expressed in the duodenum, is shed into the gut lumen and is active in LPS dephosphorylation throughout the gut lumen and in the feces. Akp3 (-/-) mice display gut dysbiosis and are more prone to dextran sodium sulfate-induced colitis than wild-type mice. Of relevance, oral administration of recombinant calf IAP prevents the dysbiosis and protects the gut from chronic colitis. Analogous to the role of IAP in the gut, TNAP expression in the liver may have a proactive role from bacterial endotoxin insult. Finally, more recent studies suggest that neuronal death in Alzheimer's disease may also be associated with TNAP function on certain brain-specific phosphoproteins. This review recounts the established roles of TNAP and IAP and briefly discusses new areas of investigation related to multisystemic functions of these isozymes.
Collapse
|
197
|
Yuan Q, Jiang Y, Zhao X, Sato T, Densmore M, Schüler C, Erben RG, McKee MD, Lanske B. Increased osteopontin contributes to inhibition of bone mineralization in FGF23-deficient mice. J Bone Miner Res 2014; 29:693-704. [PMID: 24038141 PMCID: PMC3937302 DOI: 10.1002/jbmr.2079] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 08/08/2013] [Accepted: 08/20/2013] [Indexed: 02/05/2023]
Abstract
Excessive FGF23 has been identified as a pivotal phosphaturic factor leading to renal phosphate-wasting and the subsequent development of rickets and osteomalacia. In contrast, loss of FGF23 in mice (Fgf23(-/-) ) leads to high serum phosphate, calcium, and 1,25-vitamin D levels, resulting in early lethality attributable to severe ectopic soft-tissue calcifications and organ failure. Paradoxically, Fgf23(-/-) mice exhibit a severe defect in skeletal mineralization despite high levels of systemic mineral ions and abundant ectopic mineralization, an abnormality that remains largely unexplained. Through use of in situ hybridization, immunohistochemistry, and immunogold labeling coupled with electron microscopy of bone samples, we discovered that expression and accumulation of osteopontin (Opn/OPN) was markedly increased in Fgf23(-/-) mice. These results were confirmed by qPCR analyses of Fgf23(-/-) bones and ELISA measurements of serum OPN. To investigate whether elevated OPN levels were contributing to the bone mineralization defect in Fgf23(-/-) mice, we generated Fgf23(-/-) /Opn(-/-) double-knockout mice (DKO). Biochemical analyses showed that the hypercalcemia and hyperphosphatemia observed in Fgf23(-/-) mice remained unchanged in DKO mice; however, micro-computed tomography (µCT) and histomorphometric analyses showed a significant improvement in total mineralized bone volume. The severe osteoidosis was markedly reduced and a normal mineral apposition rate was present in DKO mice, indicating that increased OPN levels in Fgf23(-/-) mice are at least in part responsible for the osteomalacia. Moreover, the increased OPN levels were significantly decreased upon lowering serum phosphate by feeding a low-phosphate diet or after deletion of NaPi2a, indicating that phosphate levels contribute in part to the high OPN levels in Fgf23(-/-) mice. In summary, our results suggest that increased OPN is an important pathogenic factor mediating the mineralization defect and the alterations in bone metabolism observed in Fgf23(-/-) bones.
Collapse
Affiliation(s)
- Quan Yuan
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, USA; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
198
|
Ryu WS, Lee SH, Kim CK, Kim BJ, Kwon HM, Yoon BW. High serum alkaline phosphatase in relation to cerebral small vessel disease. Atherosclerosis 2014; 232:313-8. [DOI: 10.1016/j.atherosclerosis.2013.11.047] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 10/09/2013] [Accepted: 11/14/2013] [Indexed: 10/26/2022]
|
199
|
Liedert A, Schinke T, Ignatius A, Amling M. The role of midkine in skeletal remodelling. Br J Pharmacol 2014; 171:870-8. [PMID: 24102259 PMCID: PMC3925025 DOI: 10.1111/bph.12412] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/02/2013] [Accepted: 09/09/2013] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Bone tissue is subjected to continuous remodelling, replacing old or damaged bone throughout life. In bone remodelling, the coordinated activities of bone-forming osteoblasts and bone-resorbing osteoclasts ensure the maintenance of bone mass and strength. In early life, the balance of these cellular activities is tightly regulated by various factors, including systemic hormones, the mechanical environment and locally released growth factors. Age-related changes in the activity of these factors in bone remodelling can result in diseases with low bone mass, such as osteoporosis. Osteoporosis is a systemic and age-related skeletal disease characterized by low bone mass and structural degeneration of bone tissue, predisposing the patient to an increased fracture risk. The growth factor midkine (Mdk) plays a key role in bone remodelling and it is expressed during bone formation and fracture repair. Using a mouse deficient in Mdk, our group have identified this protein as a negative regulator of bone formation and mechanically induced bone remodelling. Thus, specific Mdk antagonists might represent a therapeutic option for diseases characterized by low bone mass, such as osteoporosis. LINKED ARTICLES This article is part of a themed section on Midkine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-4.
Collapse
Affiliation(s)
- A Liedert
- Institute of Orthopedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | | | | | | |
Collapse
|
200
|
Abstract
Teeth are mineralized organs composed of three unique hard tissues, enamel, dentin, and cementum, and supported by the surrounding alveolar bone. Although odontogenesis differs from osteogenesis in several respects, tooth mineralization is susceptible to similar developmental failures as bone. Here we discuss conditions fitting under the umbrella of rickets, which traditionally referred to skeletal disease associated with vitamin D deficiency but has been more recently expanded to include newly identified factors involved in endocrine regulation of vitamin D, phosphate, and calcium, including phosphate-regulating endopeptidase homolog, X-linked, fibroblast growth factor 23, and dentin matrix protein 1. Systemic mineral metabolism intersects with local regulation of mineralization, and factors including tissue nonspecific alkaline phosphatase are necessary for proper mineralization, where rickets can result from loss of activity of tissue nonspecific alkaline phosphatase. Individuals suffering from rickets often bear the additional burden of a defective dentition, and transgenic mouse models have aided in understanding the nature and mechanisms involved in tooth defects, which may or may not parallel rachitic bone defects. This report reviews dental effects of the range of rachitic disorders, including discussion of etiologies of hereditary forms of rickets, a survey of resulting bone and tooth mineralization disorders, and a discussion of mechanisms, known and hypothesized, involved in the observed dental pathologies. Descriptions of human pathology are augmented by analysis of transgenic mouse models, and new interpretations are brought to bear on questions of how teeth are affected under conditions of rickets. In short, the rachitic tooth will be revealed.
Collapse
Affiliation(s)
- Brian L Foster
- National Institute for Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | | | | |
Collapse
|