1
|
Deering J, Buss DJ, Kröger R, Vali H, Lagos MJ, Reznikov N, McKee MD. Bone mineralization and the effects of elevated osteopontin: from symmetry-breaking foci to 3D space-filling tessellation. Faraday Discuss 2025. [PMID: 40439195 PMCID: PMC12123585 DOI: 10.1039/d5fd00013k] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/19/2025] [Indexed: 06/02/2025]
Abstract
At the nanoscale, lamellar bone tissue mineralization ensues via heteronucleation of small mineral foci within the osteoid. The foci grow to produce a mature, volume-filling tessellation pattern at the micrometer-scale. Mineralization-inhibiting osteopontin (OPN) mediates this bone mineralization pathway and, eventually, the microscale properties of bone tissue. Using 2D and 3D electron microscopy, here we have assessed how the abundance of OPN can affect nanoscale mineralization, mineral ripening, and microscale patterning of mineral in normal wild-type mouse bone, and we compare that to mutant mouse models having elevated OPN (Fgf23-/- and Hyp mice). When OPN is elevated, volume-filling mineral tessellation was incomplete (showing a four-fold increase in mineral surface area in the vicinity of the mineralization front in Hyp bone). Immunogold labeling showed excessive OPN in the foci, suggesting an arrest of their growth and an interruption of the pathway towards microscale tessellation. In Fgf23-/- mice, electron tomography and 3D focused ion beam-scanning electron microscopy (FIB-SEM) imaging of mineral foci show instances of core-shell morphology with crystalline mineral confined to the focus interior, and an amorphous nanogranular texture persisting in the outer shell. Electron energy-loss spectroscopy, which is sensitive to nanoscale elemental composition, showed a lower Ca/P ratio at the periphery of Hyp foci, consistent with a more amorphous mineral character, suggesting that OPN may play a role in delaying the amorphous-to-crystalline transition. These aspects of nanoscale mineral maturation in mutant mice having elevated OPN implicate this protein as a fine-tuning regulator of mineralization kinetics, mineral composition, and mechanical properties of bone.
Collapse
Affiliation(s)
- Joseph Deering
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Strathcona Anatomy and Dentistry Bldg, Rm M73 3640 University Street, Montreal, QC, H3A 0C7, Canada.
| | - Daniel J Buss
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, H3A 0C7, Canada
| | - Roland Kröger
- Department of Physics, University of York, York, YO10 5DD, UK
| | - Hojatollah Vali
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, H3A 0C7, Canada
| | - Maureen J Lagos
- Department of Materials Science and Engineering, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Natalie Reznikov
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Strathcona Anatomy and Dentistry Bldg, Rm M73 3640 University Street, Montreal, QC, H3A 0C7, Canada.
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, H3A 0C7, Canada
- Department of Bioengineering, Faculty of Engineering, McGill University, Montreal, QC, H3A 0E9, Canada
| | - Marc D McKee
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Strathcona Anatomy and Dentistry Bldg, Rm M73 3640 University Street, Montreal, QC, H3A 0C7, Canada.
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, H3A 0C7, Canada
| |
Collapse
|
2
|
Radvar E, Mehta K, D'Ambrosio A, Mastroianni G, Al-Jawad M, Stevens MM, Mata A, Elsharkawy S. Investigating the role of elastin and extracellular matrix damage in cardiovascular calcification. J Struct Biol 2025; 217:108140. [PMID: 39638017 DOI: 10.1016/j.jsb.2024.108140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Although calcification in the cardiovascular system is highly studied, the mechanisms behind it are not well understood. Current proposed mechanisms focus on cellular processes leading to, or controlling the unwanted mineralization in soft tissues. However, extracellular components such as collagen and elastin fundamentally regulate the mechanical properties of heart tissues. Here, we report on a toolkit to control the composition of tissues through the selective digestion of extracellular matrix (ECM) components, which can be used to design disease-specific in vitro models. Using this technique, we show that elastin as well as matrix tissue damage may play major role in cardiovascular calcification. This study highlights a novel approach to understand the role of proteins in soft tissue calcifications and may lead to the development of strategies to treat and prevent these unwanted pathological disorders.
Collapse
Affiliation(s)
- Elham Radvar
- Centre for Oral, Clinical, and Translational Sciences, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, United Kingdom
| | - Khushbu Mehta
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London E1 2DP, United Kingdom
| | - Alexander D'Ambrosio
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London E1 2DP, United Kingdom
| | - Giulia Mastroianni
- School of Biological and Behavioral Sciences, Queen Mary University of London, London E1 2DP, United Kingdom
| | - Maisoon Al-Jawad
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2DP, United Kingdom; School of Dentistry, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London SW7 2AZ, United Kingdom; Department of Physiology, Anatomy and Genetics, Department of Engineering Science, Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK; Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Alvaro Mata
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London E1 2DP, United Kingdom; School of Pharmacy, Department of Chemical and Environmental Engineering and Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, United Kingdom.
| | - Sherif Elsharkawy
- Centre for Oral, Clinical, and Translational Sciences, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, United Kingdom; Prosthodontics Department, Dental Directorate, Guy's and St Thomas' NHS Trust, London SE1 9RT, United Kingdom; London Centre of Nanotechnology, London WC1H 0AH, United Kingdom; Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London E1 2DP, United Kingdom; Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2DP, United Kingdom.
| |
Collapse
|
3
|
Hengtrakul N, Furrow E, Borofsky M, Toth F, Lulich JP. Expression of osteogenic proteins in kidneys of cats with nephrocalcinosis. J Vet Intern Med 2025; 39:e17278. [PMID: 39757788 PMCID: PMC11702495 DOI: 10.1111/jvim.17278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Nephrocalcinosis is a common pathological finding in cats with chronic kidney disease and nephrolithiasis. Understanding its pathogenesis may identify future therapeutic targets. HYPOTHESIS Nephrocalcinosis is associated with expression of an osteogenic phenotype. ANIMALS Kidneys with medullary mineralization were obtained from 18 cats (10 with and 8 without nephroliths) undergoing necropsy. METHODS Cross-sectional study. Microradiography and histopathology (modified von Kossa stain) were used to confirm parenchymal mineralization. Immunohistochemistry for 5 osteogenic markers was performed to determine their co-localization with nephrocalcinosis. The proportion of kidneys with stronger immunointensity in mineralized versus non-mineralized regions was analyzed using 1-tailed sign tests. The proportion of kidneys with co-localization of nephrocalcinosis and each marker was compared between kidneys with and without nephroliths using Fisher's exact tests. RESULTS Nephrocalcinosis co-localized with osteopontin immunoreactivity in all 18 cats (100%) and with osteocalcin in 12 cats (67%). Both osteogenic markers had stronger immunointensity in mineralized regions compared with non-mineralized regions. Limited co-localization was observed with other markers: bone morphogenic protein-2 in 2 kidneys (both with nephroliths) and tissue non-specific alkaline phosphatase in 1 kidney (without nephroliths); runt-related transcription factor-2 was undetected. No statistically significant differences were found in the co-localization of nephrocalcinosis with osteogenic proteins between kidneys with and without nephroliths. CONCLUSIONS AND CLINICAL IMPORTANCE Expression of osteogenic proteins in areas of nephrocalcinosis indicates that nephrocalcinosis is associated with the development of an osteogenic phenotype. Targeting these processes could offer a novel approach to prevent nephrolithiasis at its origin.
Collapse
Affiliation(s)
- Nuttha Hengtrakul
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSt PaulMinnesotaUSA
| | - Eva Furrow
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSt PaulMinnesotaUSA
| | - Michael Borofsky
- Department of Urology, Medical SchoolUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Ferenc Toth
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSt PaulMinnesotaUSA
| | - Jody P. Lulich
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSt PaulMinnesotaUSA
| |
Collapse
|
4
|
Hui ST, Gong L, Swichkow C, Blencowe M, Kaminska D, Diamante G, Pan C, Dalsania M, French SW, Magyar CE, Pajukanta P, Pihlajamäki J, Boström KI, Yang X, Lusis AJ. Role of Matrix Gla Protein in Transforming Growth Factor-β Signaling and Nonalcoholic Steatohepatitis in Mice. Cell Mol Gastroenterol Hepatol 2023; 16:943-960. [PMID: 37611662 PMCID: PMC10632746 DOI: 10.1016/j.jcmgh.2023.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic steatohepatitis (NASH) is a complex disease involving both genetic and environmental factors in its onset and progression. We analyzed NASH phenotypes in a genetically diverse cohort of mice, the Hybrid Mouse Diversity Panel, to identify genes contributing to disease susceptibility. METHODS A "systems genetics" approach, involving integration of genetic, transcriptomic, and phenotypic data, was used to identify candidate genes and pathways in a mouse model of NASH. The causal role of Matrix Gla Protein (MGP) was validated using heterozygous MGP knockout (Mgp+/-) mice. The mechanistic role of MGP in transforming growth factor-beta (TGF-β) signaling was examined in the LX-2 stellate cell line by using a loss of function approach. RESULTS Local cis-acting regulation of MGP was correlated with fibrosis, suggesting a causal role in NASH, and this was validated using loss of function experiments in 2 models of diet-induced NASH. Using single-cell RNA sequencing, Mgp was found to be primarily expressed in hepatic stellate cells and dendritic cells in mice. Knockdown of MGP expression in stellate LX-2 cells led to a blunted response to TGF-β stimulation. This was associated with reduced regulatory SMAD phosphorylation and TGF-β receptor ALK1 expression as well as increased expression of inhibitory SMAD6. Hepatic MGP expression was found to be significantly correlated with the severity of fibrosis in livers of patients with NASH, suggesting relevance to human disease. CONCLUSIONS MGP regulates liver fibrosis and TGF-β signaling in hepatic stellate cells and contributes to NASH pathogenesis.
Collapse
Affiliation(s)
- Simon T Hui
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| | - Lili Gong
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Chantle Swichkow
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Montgomery Blencowe
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California
| | - Dorota Kaminska
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Graciel Diamante
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California
| | - Calvin Pan
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Meet Dalsania
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Samuel W French
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Clara E Magyar
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Medicine, Endocrinology, and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Kristina I Boström
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California
| | - Aldons J Lusis
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
5
|
Fay LY, Kuo CH, Chang HK, Yeh MY, Chang CC, Ko CC, Tu TH, Kuo YH, Hsu WY, Hung CH, Chen CJ, Wu JC, Tsai MJ, Huang WC, Cheng H, Lee MJ. Comparative Study of the Cytokine Profiles of Serum and Tissues from Patients with the Ossification of the Posterior Longitudinal Ligament. Biomedicines 2023; 11:2021. [PMID: 37509659 PMCID: PMC10377187 DOI: 10.3390/biomedicines11072021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND The ossification of the posterior longitudinal ligament (OPLL) is one of the contributing factors leading to severe cervical spondylotic myelopathy (CSM). The mechanism causing ossification is still unclear. The current study was designed to analyze the specimens of patients with or without OPLL. METHODS The study collected 51 patients with cervical spondylosis. There were six serum samples in both the non-OPLL (NOPLL) and OPLL groups. For tissue analysis, there were seven samples in the NOPLL group and five samples in the OPLL group. The specimens of serum and tissue were analyzed by using Human Cytokine Antibody Arrays to differentiate biomarkers between the OPLL and NOPLL groups, as well as between serum and OPLL tissue. Immunohistochemical staining of the ligament tissue was undertaken for both groups. RESULTS For OPLL vs. NOPLL, the serum leptin levels are higher in the OPLL group, corroborating others' observations that it may serve as a disease marker. In the tissue, angiogenin (ANG), osteopontin (OPN), and osteopro-tegerin (OPG) are higher than they are in the OPLL group (p < 0.05). For serum vs. OPLL tissue, many chemotactic cytokines demonstrated elevated levels of MIP1 delta, MCP-1, and RANTES in the serum, while many cytokines promoting or regulating bone genesis were up-regulated in tissue (oncostatin M, FGF-9, LIF, osteopontin, osteoprotegerin, TGF-beta2), as well as the factor that inhibits osteoclastogenesis (IL-10), with very few cytokines responsible for osteoclastogenesis. Molecules promoting angiogenesis, including angiotensin, vEGF, and osteoprotegerin, are abundant in the OPLL tissue, which paves the way for robust bone growth.
Collapse
Affiliation(s)
- Li-Yu Fay
- Institute of Pharmacology, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Chao-Hung Kuo
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, and National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
| | - Hsuan-Kan Chang
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Mei-Yin Yeh
- Institute of Pharmacology, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Chih-Chang Chang
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Chin-Chu Ko
- Institute of Pharmacology, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Tsung-Hsi Tu
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Yi-Hsuan Kuo
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Wang-Yu Hsu
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Chien-Hui Hung
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Ching-Jung Chen
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Jau-Ching Wu
- Institute of Pharmacology, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - May-Jywan Tsai
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
- Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Wen-Cheng Huang
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Henrich Cheng
- Institute of Pharmacology, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St., Taipei 11217, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Taipei 11217, Taiwan
| | - Meng-Jen Lee
- Department of Applied Chemistry, Chaoyang University of Technology, 168, Jifeng E. Rd., Taichung 413310, Taiwan
| |
Collapse
|
6
|
Alkaissi H, McFarlane SI. Hyperhomocysteinemia and Accelerated Aging: The Pathogenic Role of Increased Homocysteine in Atherosclerosis, Osteoporosis, and Neurodegeneration. Cureus 2023; 15:e42259. [PMID: 37605676 PMCID: PMC10440097 DOI: 10.7759/cureus.42259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2023] [Indexed: 08/23/2023] Open
Abstract
Cardiovascular diseases and osteoporosis, seemingly unrelated disorders that occur with advanced age, share major pathogenetic mechanisms contributing to accelerated atherosclerosis and bone loss. Hyperhomocysteinemia (hHcy) is among these mechanisms that can cause both vascular and bone disease. In its more severe form, hHcy can present early in life as homocystinuria, an inborn error of metabolic pathways of the sulfur-containing amino acid methionine. In its milder forms, hHcy may go undiagnosed and untreated into adulthood. As such, hHcy may serve as a potential therapeutic target for cardiovascular disease, osteoporosis, thrombophilia, and neurodegeneration, collectively representing accelerated aging. Multiple trials to lower cardiovascular risk and improve bone density with homocysteine-lowering agents, yet none has proven to be clinically meaningful. To understand this unmet clinical need, this review will provide mechanistic insight into the pathogenesis of vascular and bone disease in hHcy, using homocystinuria as a model for accelerated atherosclerosis and bone density loss, a model for accelerated aging.
Collapse
Affiliation(s)
- Hussam Alkaissi
- Internal Medicine, Kings County Hospital Center, Brooklyn, USA
- Internal Medicine, Veterans Affairs Medical Center, Brooklyn, USA
- Internal Medicine, State University of New York Downstate Medical Center, Brooklyn, USA
| | - Samy I McFarlane
- Endocrinology, State University of New York Downstate Medical Center, Brooklyn, USA
| |
Collapse
|
7
|
Activating BK channels ameliorates vascular smooth muscle calcification through Akt signaling. Acta Pharmacol Sin 2022; 43:624-633. [PMID: 34163023 PMCID: PMC8888620 DOI: 10.1038/s41401-021-00704-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 05/23/2021] [Indexed: 02/06/2023]
Abstract
Vascular calcification (VC) is characterized by pathological depositions of calcium and phosphate in the arteries and veins via an active cell-regulated process, in which vascular smooth muscle cells (VSMCs) transform into osteoblast/chondrocyte-like cells as in bone formation. VC is associated with significant morbidity and mortality in chronic kidney disease (CKD) and cardiovascular disease, but the underlying mechanisms remain unclear. In this study we investigated the role of large-conductance calcium-activated potassium (BK) channels in 3 experimental VC models. VC was induced in vascular smooth muscle cells (VSMCs) by β-glycerophosphate (β-GP), or in rats by subtotal nephrectomy, or in mice by high-dosage vitamin D3. We showed that the expression of BK channels in the artery of CKD rats with VC and in β-GP-treated VSMCs was significantly decreased, which was functionally confirmed by patch-clamp recording. In β-GP-treated VSMCs, BK channel opener NS1619 (20 μM) significantly alleviated VC by decreasing calcium content and alkaline phosphatase activity. Furthermore, NS1619 decreased mRNA expression of ostoegenic genes OCN and OPN, as well as Runx2 (a key transcription factor involved in preosteoblast to osteoblast differentiation), and increased the expression of α-SMA protein, whereas BK channel inhibitor paxilline (10 μM) caused the opposite effects. In primary cultured VSMCs from BK-/- mice, BK deficiency aggravated calcification as did BK channel inhibitor in normal VSMCs. Moreover, calcification was more severe in thoracic aorta rings of BK-/- mice than in those of wild-type littermates. Administration of BK channel activator BMS191011 (10 mg· kg-1 ·d-1) in high-dosage vitamin D3-treated mice significantly ameliorated calcification. Finally, co-treatment with Akt inhibitor MK2206 (1 μM) or FoxO1 inhibitor AS1842856 (3 μM) in calcified VSMCs abrogated the effects of BK channel opener NS1619. Taken together, activation of BK channels ameliorates VC via Akt/FoxO1 signaling pathways. Strategies to activate BK channels and/or enhance BK channel expression may offer therapeutic avenues to control VC.
Collapse
|
8
|
The Roles of Osteopontin in the Pathogenesis of West Nile Encephalitis. Vaccines (Basel) 2020; 8:vaccines8040748. [PMID: 33317005 PMCID: PMC7768535 DOI: 10.3390/vaccines8040748] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/03/2020] [Accepted: 12/06/2020] [Indexed: 12/18/2022] Open
Abstract
Osteopontin (OPN), a multifunctional protein encoded by the secreted phosphoprotein-1 (Spp-1) gene in humans, plays important roles in a variety of physiological conditions, such as biomineralization, bone remodeling and immune functions. OPN also has significant roles in the pathogenesis of autoimmune, allergy and inflammatory diseases, as well as bacterial, fungal and viral infections. West Nile virus (WNV), a mosquito-transmitted flavivirus, is the leading agent for viral encephalitis in North America. Recent progress has been made in understanding both the biological functions of OPN and the pathogenesis of WNV. In this review article, we have summarized the current understanding of the biology of OPN and its vital roles in the pathogenesis of WNV encephalitis.
Collapse
|
9
|
Wang Y, Morsali R, Dai Z, Minary-Jolandan M, Qian D. Computational Nanomechanics of Noncollagenous Interfibrillar Interface in Bone. ACS APPLIED MATERIALS & INTERFACES 2020; 12:25363-25373. [PMID: 32407068 DOI: 10.1021/acsami.0c01613] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The noncollagenous interfibrillar interface in bone provides the critical function of transferring loads among collagen fibrils and their bundles, with adhesive mechanisms at this site thus significantly contributing to the mechanical properties of bone. Motivated by the experimental observations and hypotheses, a computational study is presented to elucidate the critical roles of two major proteins at the nanoscale interfibrillar interface, that is, osteopontin (OPN) and osteocalcin (OC) in bone. This study reveals the extremely high interfacial toughness of the OPN/OC composite. The previously proposed hypothesis of sacrificial bonds in the extracellular organic matrix is tested, and the remarkable mechanical properties of the nanoscale bone interface are attributed to the collaborative interactions between the OPN and OC proteins.
Collapse
Affiliation(s)
- Yang Wang
- Department of Mechanical Engineering, The University of Texas at Dallas, 800 West Campbell Road, Richardson, Texas 75080, United States
| | - Reza Morsali
- Department of Mechanical Engineering, The University of Texas at Dallas, 800 West Campbell Road, Richardson, Texas 75080, United States
| | - Zhengwei Dai
- College of Material and Textile Engineering, Jiaxing University, Jiaxing 314001, People's Republic of China
| | - Majid Minary-Jolandan
- Department of Mechanical Engineering, The University of Texas at Dallas, 800 West Campbell Road, Richardson, Texas 75080, United States
| | - Dong Qian
- Department of Mechanical Engineering, The University of Texas at Dallas, 800 West Campbell Road, Richardson, Texas 75080, United States
| |
Collapse
|
10
|
Rashdan NA, Sim AM, Cui L, Phadwal K, Roberts FL, Carter R, Ozdemir DD, Hohenstein P, Hung J, Kaczynski J, Newby DE, Baker AH, Karsenty G, Morton NM, MacRae VE. Osteocalcin Regulates Arterial Calcification Via Altered Wnt Signaling and Glucose Metabolism. J Bone Miner Res 2020; 35:357-367. [PMID: 31596966 DOI: 10.1002/jbmr.3888] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 09/23/2019] [Accepted: 09/28/2019] [Indexed: 12/12/2022]
Abstract
Arterial calcification is an important hallmark of cardiovascular disease and shares many similarities with skeletal mineralization. The bone-specific protein osteocalcin (OCN) is an established marker of vascular smooth muscle cell (VSMC) osteochondrogenic transdifferentiation and a known regulator of glucose metabolism. However, the role of OCN in controlling arterial calcification is unclear. We hypothesized that OCN regulates calcification in VSMCs and sought to identify the underpinning signaling pathways. Immunohistochemistry revealed OCN co-localization with VSMC calcification in human calcified carotid artery plaques. Additionally, 3 mM phosphate treatment stimulated OCN mRNA expression in cultured VSMCs (1.72-fold, p < 0.001). Phosphate-induced calcification was blunted in VSMCs derived from OCN null mice (Ocn -/- ) compared with cells derived from wild-type (WT) mice (0.37-fold, p < 0.001). Ocn -/- VSMCs showed reduced mRNA expression of the osteogenic marker Runx2 (0.51-fold, p < 0.01) and the sodium-dependent phosphate transporter, PiT1 (0.70-fold, p < 0.001), with an increase in the calcification inhibitor Mgp (1.42-fold, p < 0.05) compared with WT. Ocn -/- VSMCs also showed reduced mRNA expression of Axin2 (0.13-fold, p < 0.001) and Cyclin D (0.71 fold, p < 0.01), markers of Wnt signaling. CHIR99021 (GSK3β inhibitor) treatment increased calcium deposition in WT and Ocn -/- VSMCs (1 μM, p < 0.001). Ocn -/- VSMCs, however, calcified less than WT cells (1 μM; 0.27-fold, p < 0.001). Ocn -/- VSMCs showed reduced mRNA expression of Glut1 (0.78-fold, p < 0.001), Hex1 (0.77-fold, p < 0.01), and Pdk4 (0.47-fold, p < 0.001). This was accompanied by reduced glucose uptake (0.38-fold, p < 0.05). Subsequent mitochondrial function assessment revealed increased ATP-linked respiration (1.29-fold, p < 0.05), spare respiratory capacity (1.59-fold, p < 0.01), and maximal respiration (1.52-fold, p < 0.001) in Ocn -/- versus WT VSMCs. Together these data suggest that OCN plays a crucial role in arterial calcification mediated by Wnt/β-catenin signaling through reduced maximal respiration. Mitochondrial dynamics may therefore represent a novel therapeutic target for clinical intervention. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Nabil A Rashdan
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Alisia M Sim
- School of Chemistry, University of Edinburgh, Edinburgh, UK
| | - Lin Cui
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Kanchan Phadwal
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Fiona L Roberts
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Roderick Carter
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Derya D Ozdemir
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Peter Hohenstein
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - John Hung
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Jakub Kaczynski
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - David E Newby
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Andrew H Baker
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Gerard Karsenty
- Department of Genetics and Development, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Nicholas M Morton
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Vicky E MacRae
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
11
|
Bäck M, Aranyi T, Cancela ML, Carracedo M, Conceição N, Leftheriotis G, Macrae V, Martin L, Nitschke Y, Pasch A, Quaglino D, Rutsch F, Shanahan C, Sorribas V, Szeri F, Valdivielso P, Vanakker O, Kempf H. Endogenous Calcification Inhibitors in the Prevention of Vascular Calcification: A Consensus Statement From the COST Action EuroSoftCalcNet. Front Cardiovasc Med 2019; 5:196. [PMID: 30713844 PMCID: PMC6345677 DOI: 10.3389/fcvm.2018.00196] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/19/2018] [Indexed: 01/29/2023] Open
Abstract
The physicochemical deposition of calcium-phosphate in the arterial wall is prevented by calcification inhibitors. Studies in cohorts of patients with rare genetic diseases have shed light on the consequences of loss-of-function mutations for different calcification inhibitors, and genetic targeting of these pathways in mice have generated a clearer picture on the mechanisms involved. For example, generalized arterial calcification of infancy (GACI) is caused by mutations in the enzyme ecto-nucleotide pyrophosphatase/phosphodiesterase-1 (eNPP1), preventing the hydrolysis of ATP into pyrophosphate (PPi). The importance of PPi for inhibiting arterial calcification has been reinforced by the protective effects of PPi in various mouse models displaying ectopic calcifications. Besides PPi, Matrix Gla Protein (MGP) has been shown to be another potent calcification inhibitor as Keutel patients carrying a mutation in the encoding gene or Mgp-deficient mice develop spontaneous calcification of the arterial media. Whereas PPi and MGP represent locally produced calcification inhibitors, also systemic factors contribute to protection against arterial calcification. One such example is Fetuin-A, which is mainly produced in the liver and which forms calciprotein particles (CPPs), inhibiting growth of calcium-phosphate crystals in the blood and thereby preventing their soft tissue deposition. Other calcification inhibitors with potential importance for arterial calcification include osteoprotegerin, osteopontin, and klotho. The aim of the present review is to outline the latest insights into how different calcification inhibitors prevent arterial calcification both under physiological conditions and in the case of disturbed calcium-phosphate balance, and to provide a consensus statement on their potential therapeutic role for arterial calcification.
Collapse
Affiliation(s)
- Magnus Bäck
- Translational Cardiology, Center for Molecular Medicine, Karolinska University Hospital Stockholmt, Stockholm, Sweden
| | - Tamas Aranyi
- Research Center for Natural Sciences, Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary
| | - M Leonor Cancela
- Department of Biomedical Sciences and Medicine, Algarve Biomedical Centre, Centre of Marine Sciences/CCMAR, University of Algarve, Faro, Portugal
| | - Miguel Carracedo
- Translational Cardiology, Center for Molecular Medicine, Karolinska University Hospital Stockholmt, Stockholm, Sweden
| | - Natércia Conceição
- Department of Biomedical Sciences and Medicine, Algarve Biomedical Centre, Centre of Marine Sciences/CCMAR, University of Algarve, Faro, Portugal
| | - Georges Leftheriotis
- LP2M, University of Nice-Sophia Antipolis and Vascular Physiology and Medicine, University Hospital of Nice, Nice, France
| | - Vicky Macrae
- The Roslin Institute and Royal School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Ludovic Martin
- PXE Reference Center, Angers University Hospital, Angers, France
| | - Yvonne Nitschke
- Department of General Pediatrics, Münster University Children's Hospital, Münster, Germany
| | | | - Daniela Quaglino
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Frank Rutsch
- Department of General Pediatrics, Münster University Children's Hospital, Münster, Germany
| | - Catherine Shanahan
- British Heart Foundation Centre of Research Excellence, James Black Centre, School of Cardiovascular Medicine and Sciences, King's College London, London, United Kingdom
| | - Victor Sorribas
- Laboratory of Molecular Toxicology, Veterinary Faculty, University of Zaragoza, Zaragoza, Spain
| | - Flora Szeri
- Research Center for Natural Sciences, Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary.,Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Pedro Valdivielso
- Internal Medicine, Instituto de Investigación Biomédica (IBIMA), Virgen de la Victoria University Hospital, Universidad de Málaga, Málaga, Spain
| | - Olivier Vanakker
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Hervé Kempf
- UMR 7365 CNRS-Université de Lorraine, IMoPA, Vandoeuvre-lès-Nancy, France
| |
Collapse
|
12
|
Hoac B, Nelea V, Jiang W, Kaartinen MT, McKee MD. Mineralization-inhibiting effects of transglutaminase-crosslinked polymeric osteopontin. Bone 2017; 101:37-48. [PMID: 28428079 DOI: 10.1016/j.bone.2017.04.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 03/21/2017] [Accepted: 04/16/2017] [Indexed: 01/12/2023]
Abstract
Osteopontin (OPN) belongs to the SIBLING family (Small, Integrin-Binding LIgand N-linked Glycoproteins) of mineral-binding matrix proteins found in bones and teeth. OPN is a well-known inhibitor of matrix mineralization, and enzymatic modification of OPN can affect this inhibitory function. In bone, OPN exists both as a monomer and as a high-molecular-weight polymer - the latter is formed by transglutaminase-mediated crosslinking of glutamine and lysine residues in OPN to create homotypic protein assemblies. OPN can be covalently crosslinked by transglutaminase 2 (TG2) and Factor XIII-A. Polymeric OPN has increased binding to collagen and promotes osteoblast adhesion, but despite these initial observations, its role in mineralization is not clear. In this study, we investigated the effect of polymerized OPN on mineralization using a hydroxyapatite crystal growth assay and mineralizing MC3T3-E1 osteoblast cultures. In the cultures, endogenous polymeric OPN was detected after mineralization occurred. In cell-free conditions, TG2 was used to crosslink bovine OPN into its polymeric form, and atomic force microscopy and dynamic light scattering revealed variably-sized, large branched aggregates ranging across hundreds of nanometers. These OPN polymers inhibited the growth of hydroxyapatite crystals in solution at concentrations similar to monomeric OPN, although the crosslinking slightly reduced its inhibitory potency. When added to MC3T3-E1 osteoblast cultures, this exogenous polymeric OPN essentially did not inhibit mineralization when given during the later mineralization stages of culture; however, cultures treated early and then continuously with polymeric OPN throughout both the matrix assembly and mineral deposition stages showed reduced mineralization. Immunoblotting of protein extracts from these continuously treated cultures revealed exogenous OPN polymers incorporated into mature matrix that had not yet mineralized. These results suggest that in bone, the increased size and branched structure of crosslinked inhibitory polymeric OPN near the mineralization front could hinder it from accessing focal mineralization sites in the dense collagen-rich matrix, suggesting that OPN-crosslinking into polymers may represent a way to fine-tune the inhibitory potency of OPN on bone mineralization.
Collapse
Affiliation(s)
- Betty Hoac
- Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - Valentin Nelea
- Faculty of Dentistry, McGill University, Montreal, QC, Canada; Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Wenge Jiang
- Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - Mari T Kaartinen
- Faculty of Dentistry, McGill University, Montreal, QC, Canada; Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Marc D McKee
- Faculty of Dentistry, McGill University, Montreal, QC, Canada; Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
13
|
Borrás T. A single gene connects stiffness in glaucoma and the vascular system. Exp Eye Res 2017; 158:13-22. [PMID: 27593913 PMCID: PMC6067113 DOI: 10.1016/j.exer.2016.08.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/26/2016] [Accepted: 08/31/2016] [Indexed: 12/13/2022]
Abstract
Arterial calcification results in arterial stiffness and higher systolic blood pressure. Arterial calcification is prevented by the high expression of the Matrix-Gla gene (MGP) in the vascular smooth muscle cells (VSMC) of the arteries' tunica media. Originally, MGP, a gene highly expressed in cartilage and VSMC, was found to be one of the top expressed genes in the trabecular meshwork. The creation of an Mgp-lacZ Knock-In mouse and the use of mouse genetics revealed that in the eye, Mgp's abundant expression is localized and restricted to glaucoma-associated tissues from the anterior and posterior segments. In particular, it is specifically expressed in the regions of the trabecular meshwork and of the peripapillary sclera that surrounds the optic nerve. Because stiffness in these tissues would significantly alter outflow facility and biomechanical scleral stress in the optic nerve head (ONH), we propose MGP as a strong candidate for the regulation of stiffness in glaucoma. MGP further illustrates the presence of a common function affecting key glaucomatous parameters in the front and back of the eye, and thus offers the possibility for a sole therapeutic target for the disease.
Collapse
Affiliation(s)
- Teresa Borrás
- Department of Ophthalmology, University of North Carolina School of Medicine, 4109C Neuroscience Research Building CB 7041, 105 Mason Farm Road, Chapel Hill, NC 27599-7041, USA.
| |
Collapse
|
14
|
Higgins CL, Isbilir S, Basto P, Chen IY, Vaduganathan M, Vaduganathan P, Reardon MJ, Lawrie G, Peterson L, Morrisett JD. Distribution of alkaline phosphatase, osteopontin, RANK ligand and osteoprotegerin in calcified human carotid atheroma. Protein J 2016; 34:315-28. [PMID: 26307009 DOI: 10.1007/s10930-015-9620-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Ectopic vascular calcification is a significant component of atherosclerotic disease. Osteopontin (OPN), Osteoprotegerin (OPG), Receptor Activator of NFκB Ligand (RANKL), and alkaline phosphatase (ALP) are each thought to play central roles in the calcification or demineralization of atherosclerotic lesions. Abnormalities in the balance of these proteins may lead to perturbations in bone remodeling and arterial calcification. The purpose of this study was to measure the distribution of these proteins in human carotid lesions and to elucidate possible mechanism(s) whereby they control the deposition or depletion of arterial calcification. Thirty-three patients who had undergone carotid endarterectomy (CEA) within the previous 18 months and 11 control patients were enrolled. CEA specimens were analyzed by EBCT for calcification content in terms of Agatston (AGAT) and Volume scores. CEA specimens were then cut into 5 mm segments which were homogenized and extracted. Extracts were analyzed for tissue levels of calcium, phosphorus, ALP, OPN, RANKL, and OPG. Fasting blood samples were analyzed for the same components. In CEA tissue segments, the calcification levels (CHA AGAT) were inversely associated with the levels of OPG (r = -0.432/-0.579, p < 0.05) and positively associated with the levels of RANKL (r = 0.332/0.415, p < 0.05). In turn, the tissue levels of OPG were associated with homologous serum levels of OPG (r = 0.820/0.389, p < 0.001), and the tissue levels of RANKL were associated with the serum levels of homologous RANKL (r = 0.739/0.666, p < 0.0001). This study suggests that serum levels of OPG and RANKL may be useful biomarkers for estimating the degree of calcification in carotid atherosclerotic lesions.
Collapse
Affiliation(s)
| | - Salim Isbilir
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Pamela Basto
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Iou Yih Chen
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Muthiah Vaduganathan
- Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital, A601, 6565 Fannin St., Houston, TX, 77030, USA
| | - Periyanan Vaduganathan
- Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital, A601, 6565 Fannin St., Houston, TX, 77030, USA
| | - Michael J Reardon
- Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital, A601, 6565 Fannin St., Houston, TX, 77030, USA
| | - Gerald Lawrie
- Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital, A601, 6565 Fannin St., Houston, TX, 77030, USA
| | - Leif Peterson
- Department of Public Health, The Methodist Hospital Research Institute, Houston, TX, USA
| | - Joel D Morrisett
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA. .,Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital, A601, 6565 Fannin St., Houston, TX, 77030, USA.
| |
Collapse
|
15
|
Paloian NJ, Leaf EM, Giachelli CM. Osteopontin protects against high phosphate-induced nephrocalcinosis and vascular calcification. Kidney Int 2016; 89:1027-1036. [PMID: 27083280 DOI: 10.1016/j.kint.2015.12.046] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 12/08/2015] [Accepted: 12/30/2015] [Indexed: 01/09/2023]
Abstract
Pathologic calcification is a significant cause of increased morbidity and mortality in patients with chronic kidney disease. The precise mechanisms of ectopic calcification are not fully elucidated, but it is known to be caused by an imbalance of procalcific and anticalcific factors. In the chronic kidney disease population, an elevated phosphate burden is both highly prevalent and a known risk factor for ectopic calcification. Here we tested whether osteopontin, an inhibitor of calcification, protects against high phosphate load-induced nephrocalcinosis and vascular calcification. Osteopontin knockout mice were placed on a high phosphate diet for 11 weeks. Osteopontin deficiency together with phosphate overload caused uremia, nephrocalcinosis characterized by substantial renal tubular and interstitial calcium deposition, and marked vascular calcification when compared with control mice. Although the osteopontin-deficient mice did not exhibit hypercalcemia or hyperphosphatemia, they did show abnormalities in the mineral metabolism hormone fibroblast growth factor-23. Thus, endogenous osteopontin plays a critical role in the prevention of phosphate-induced nephrocalcinosis and vascular calcification in response to high phosphate load. A better understanding of osteopontin's role in phosphate-induced calcification will hopefully lead to better biomarkers and therapies for this disease, especially in patients with chronic kidney disease and other at-risk populations.
Collapse
Affiliation(s)
- Neil J Paloian
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Elizabeth M Leaf
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Cecilia M Giachelli
- Department of Bioengineering, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
16
|
Avogaro A, Fadini GP. Mechanisms of ectopic calcification: implications for diabetic vasculopathy. Cardiovasc Diagn Ther 2015; 5:343-52. [PMID: 26543821 DOI: 10.3978/j.issn.2223-3652.2015.06.05] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Vascular calcification (VC) is the deposition of calcium/phosphate in the vasculature, which portends a worse clinical outcome and predicts major adverse cardiovascular events. VC is an active process initiated and regulated via a variety of molecular signalling pathways. There are mainly two types of calcifications: the media VC and the intima VC. All major risk factors for cardiovascular disease (CVD) have been linked to the presence/development of VC. Besides the risk factors, a genetic component is also operative to determine arterial calcification. Several events take place before VC is established, including inflammation, trans-differentiation of vascular cells and homing of circulating pro-calcific cells. Diabetes is an important predisposing factor for VC. Compared with non-diabetic subjects, patients with diabetes show increased VC and higher expression of bone-related proteins in the medial layer of the vessels. In this review we will highlight the mechanisms underlying vascular calcification in diabetic patients.
Collapse
Affiliation(s)
- Angelo Avogaro
- 1 Division of Metabolic Diseases, Department of Medicine, University of Padova, Padova, Italy ; 2 Laboratory of Experimental Diabetology, Venetian Institute of Molecular Medicine, Padova, Italy
| | - Gian Paolo Fadini
- 1 Division of Metabolic Diseases, Department of Medicine, University of Padova, Padova, Italy ; 2 Laboratory of Experimental Diabetology, Venetian Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
17
|
Borrás T, Smith MH, Buie LK. A Novel Mgp-Cre Knock-In Mouse Reveals an Anticalcification/Antistiffness Candidate Gene in the Trabecular Meshwork and Peripapillary Scleral Region. Invest Ophthalmol Vis Sci 2015; 56:2203-14. [PMID: 25711639 PMCID: PMC4406033 DOI: 10.1167/iovs.15-16460] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 02/14/2015] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Soft tissue calcification is a pathological condition. Matrix Gla (MGP) is a potent mineralization inhibitor secreted by cartilage chondrocytes and arteries' vascular smooth muscle cells. Mgp knock-out mice die at 6 weeks due to massive arterial calcification. Arterial calcification results in arterial stiffness and higher systolic blood pressure. Intriguingly, MGP was highly abundant in trabecular meshwork (TM). Because tissue stiffness is relevant to glaucoma, we investigated which additional eye tissues use Mgp's function using knock-in mice. METHODS An Mgp-Cre-recombinase coding sequence (Cre) knock-in mouse, containing Mgp DNA plus an internal ribosomal entry site (IRES)-Cre-cassette was generated by homologous recombination. Founders were crossed with Cre-mediated reporter mouse R26R-lacZ. Their offspring expresses lacZ where Mgp is transcribed. Eyes from MgpCre/+;R26RlacZ/+ (Mgp-lacZ knock-in) and controls, 1 to 8 months were assayed for β-gal enzyme histochemistry. RESULTS As expected, Mgp-lacZ knock-in's TM was intensely blue. In addition, this mouse revealed high specific expression in the sclera, particularly in the peripapillary scleral region (ppSC). Ciliary muscle and sclera above the TM were also positive. Scleral staining was located immediately underneath the choroid (chondrocyte layer), began midsclera and was remarkably high in the ppSC. Cornea, iris, lens, ciliary body, and retina were negative. All mice exhibited similar staining patterns. All controls were negative. CONCLUSIONS Matrix Gla's restricted expression to glaucoma-associated tissues from anterior and posterior segments suggests its involvement in the development of the disease. Matrix Gla's anticalcification/antistiffness properties in the vascular tissue, together with its high TM and ppCS expression, place this gene as a strong candidate for TM's softness and sclera's stiffness regulation in glaucoma.
Collapse
Affiliation(s)
- Teresa Borrás
- Department of Ophthalmology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States
| | - Matthew H. Smith
- Department of Ophthalmology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States
| | - LaKisha K. Buie
- Department of Ophthalmology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States
| |
Collapse
|
18
|
Identification of transglutaminase reactive residues in human osteopontin and their role in polymerization. PLoS One 2014; 9:e113650. [PMID: 25419572 PMCID: PMC4242673 DOI: 10.1371/journal.pone.0113650] [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: 08/27/2014] [Accepted: 10/26/2014] [Indexed: 01/21/2023] Open
Abstract
Osteopontin (OPN) is a highly posttranslationally modified protein present in several tissues where it is implicated in numerous physiological processes. OPN primarily exerts its functions through interaction with integrins via the Arg-Gly-Asp and Ser-Val-Val-Tyr-Gly-Leu-Arg sequences located in the N-terminal part of the protein. OPN can be polymerized by the cross-linking enzyme transglutaminase 2 (TG2), and polymerization has been shown to enhance the biological activity of OPN. However, little is known about the reactivity and location of the glutamine and lysine residues involved in the TG2-mediated modification of OPN. Here we show that TG2 catalyses the incorporation of 5-(Biotinamido)pentylamine at glutamines in both the N- and C-terminal parts of OPN, whereas TG2 primarily incorporated the glutamine-donor peptide biotinyl-TVQQEL-OH into the C-terminal part of OPN. By mass spectrometric analyses we identified Gln34, Gln42, Gln193 and Gln248 as the major TG2 reactive glutamines in OPN. The distribution of reactive Gln and Lys residues in OPN proved to be important, as the full-length protein but not the physiologically highly active integrin-binding N-terminal part of OPN were able to polymerize in a TG2-mediated reaction. Collectively, these data provide important new molecular knowledge about the mechanism of OPN polymerization.
Collapse
|
19
|
Paloian NJ, Giachelli CM. A current understanding of vascular calcification in CKD. Am J Physiol Renal Physiol 2014; 307:F891-900. [PMID: 25143458 DOI: 10.1152/ajprenal.00163.2014] [Citation(s) in RCA: 246] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Patients with chronic kidney disease (CKD) and end-stage renal disease (ESRD) have significant cardiovascular morbidity and mortality that is in part due to the development of vascular calcification. Vascular calcification is an active, highly regulated process that shares many similarities with normal bone formation. New discoveries related to extracellular vesicles, microRNAs, and calciprotein particles continue to reveal the mechanisms that are involved in the initiation and progression of vascular calcification in CKD. Further innovations in these fields are critical for the development of biomarkers and therapeutic options for patients with CKD and ESRD.
Collapse
Affiliation(s)
- Neil J Paloian
- Division of Nephrology, Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington; and
| | | |
Collapse
|
20
|
Wolak T. Osteopontin - a multi-modal marker and mediator in atherosclerotic vascular disease. Atherosclerosis 2014; 236:327-37. [PMID: 25128758 DOI: 10.1016/j.atherosclerosis.2014.07.004] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/26/2014] [Accepted: 07/07/2014] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a chronic inflammatory process of the vessel wall with systemic correlates. It is now well established that patients' outcome is tightly linked to atherosclerotic plaque stability, potentially more so than to the mere plaque size. Osteopontin (OPN) is an integrin-binding ligand, N-linked glycoprotein, which was recognized as a significant participant in the atherosclerotic inflammatory milieu. Evidence from several genetic mouse models suggests that OPN is an enhancer of atherosclerosis. This may be mediated by its capacity to enhance inflammation in the atherosclerotic plaque. Interestingly, OPN may also possess potentially protective vascular effects, such as attenuation of vascular calcification. In humans circulating levels of OPN were found to be independently associated with the severity of coronary atherosclerosis. Moreover, several studies report that high plasma OPN levels were associated with increased risk for major adverse cardiac events. This review aims to critically assess current understanding of the role of OPN in the atherosclerotic process, from animal models to clinical practice. Specific focus is given to evaluating whether OPN could serve as a marker for monitoring coronary atherosclerosis severity, and in parallel, assess the evidence for its role as a mediator in the pathogenic pathways leading to atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Talya Wolak
- Hypertension Unit Faculty of Health Sciences, Soroka University Medical Center, Ben-Gurion University of the Negev, Be'er-Sheva, Israel.
| |
Collapse
|
21
|
Khavandgar Z, Roman H, Li J, Lee S, Vali H, Brinckmann J, Davis EC, Murshed M. Elastin haploinsufficiency impedes the progression of arterial calcification in MGP-deficient mice. J Bone Miner Res 2014; 29:327-37. [PMID: 23857752 DOI: 10.1002/jbmr.2039] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 05/18/2013] [Accepted: 06/10/2013] [Indexed: 01/12/2023]
Abstract
Matrix gla protein (MGP) is a potent inhibitor of extracellular matrix (ECM) mineralization. MGP-deficiency in humans leads to Keutel syndrome, a rare genetic disease hallmarked by abnormal soft tissue calcification. MGP-deficient (Mgp(-/-)) mice show progressive deposition of hydroxyapatite minerals in the arterial walls and die within 2 months of age. The mechanism of antimineralization function of MGP is not fully understood. We examined the progression of vascular calcification and expression of several chondrogenic/osteogenic markers in the thoracic aortas of Mgp(-/-) mice at various ages. Although cells with chondrocyte-like morphology have been reported in the calcified aorta, our gene expression data indicate that chondrogenic/osteogenic markers are not upregulated in the arteries prior to the initiation of calcification. Interestingly, arterial calcification in Mgp(-/-) mice appears first in the elastic laminae. Considering the known mineral scaffolding function of elastin (ELN), a major elastic lamina protein, we hypothesize that elastin content in the laminae is a critical determinant for arterial calcification in Mgp(-/-) mice. To investigate this, we performed micro-computed tomography (µCT) and histological analyses of the aortas of Mgp(-/-);Eln(+/-) mice and show that elastin haploinsufficiency significantly reduces arterial calcification in this strain. Our data suggest that MGP deficiency leads to alterations of vascular ECM that may in turn initiate arterial calcification.
Collapse
|
22
|
Park HC, Champakalakshmi R, Panengad PP, Raghunath M, Mehta JS. Tissue adhesives in ocular surgery. EXPERT REVIEW OF OPHTHALMOLOGY 2014. [DOI: 10.1586/eop.11.64] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
23
|
Beazley KE, Reckard S, Nurminsky D, Lima F, Nurminskaya M. Two sides of MGP null arterial disease: chondrogenic lesions dependent on transglutaminase 2 and elastin fragmentation associated with induction of adipsin. J Biol Chem 2013; 288:31400-8. [PMID: 24036114 PMCID: PMC3829453 DOI: 10.1074/jbc.m113.495556] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 09/02/2013] [Indexed: 01/04/2023] Open
Abstract
Mutations in matrix Gla protein (MGP) have been correlated with vascular calcification. In the mouse model, MGP null vascular disease presents as calcifying cartilaginous lesions and mineral deposition along elastin lamellae (elastocalcinosis). Here we examined the mechanisms underlying both of these manifestations. Genetic ablation of enzyme transglutaminase 2 (TG2) in Mgp(-/-) mice dramatically reduced the size of cartilaginous lesions in the aortic media, attenuated calcium accrual more than 2-fold, and doubled longevity as compared with control Mgp(-/-) animals. Nonetheless, the Mgp(-/-);Tgm2(-/-) mice still died prematurely as compared with wild-type and retained the elastocalcinosis phenotype. This pathology in Mgp(-/-) animals was developmentally preceded by extensive fragmentation of elastic lamellae and associated with elevated serine elastase activity in aortic tissue and vascular smooth muscle cells. Systematic gene expression analysis followed by an immunoprecipitation study identified adipsin as the major elastase that is induced in the Mgp(-/-) vascular smooth muscle even in the TG2 null background. These results reveal a central role for TG2 in chondrogenic transformation of vascular smooth muscle and implicate adipsin in elastin fragmentation and ensuing elastocalcinosis. The importance of elastin calcification in MGP null vascular disease is highlighted by significant residual vascular calcification and mortality in Mgp(-/-);Tgm2(-/-) mice with reduced cartilaginous lesions. Our studies identify two potential therapeutic targets in vascular calcification associated with MGP dysfunction and emphasize the need for a comprehensive approach to this multifaceted disorder.
Collapse
Affiliation(s)
- Kelly E. Beazley
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Steven Reckard
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Dmitry Nurminsky
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Florence Lima
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Maria Nurminskaya
- From the Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| |
Collapse
|
24
|
Beazley KE, Lima F, Borras T, Nurminskaya M. Attenuation of chondrogenic transformation in vascular smooth muscle by dietary quercetin in the MGP-deficient mouse model. PLoS One 2013; 8:e76210. [PMID: 24098781 PMCID: PMC3786963 DOI: 10.1371/journal.pone.0076210] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 08/21/2013] [Indexed: 12/30/2022] Open
Abstract
RATIONALE Cartilaginous metaplasia of vascular smooth muscle (VSM) is characteristic for arterial calcification in diabetes and uremia and in the background of genetic alterations in matrix Gla protein (MGP). A better understanding of the molecular details of this process is critical for the development of novel therapeutic approaches to VSM transformation and arterial calcification. OBJECTIVE This study aimed to identify the effects of bioflavonoid quercetin on chondrogenic transformation and calcification of VSM in the MGP-null mouse model and upon TGF-β3 stimulation in vitro, and to characterize the associated alterations in cell signaling. METHODS AND RESULTS Molecular analysis revealed activation of β-catenin signaling in cartilaginous metaplasia in Mgp-/- aortae in vivo and during chondrogenic transformation of VSMCs in vitro. Quercetin intercepted chondrogenic transformation of VSM and blocked activation of β-catenin both in vivo and in vitro. Although dietary quercetin drastically attenuated calcifying cartilaginous metaplasia in Mgp-/- animals, approximately one-half of total vascular calcium mineral remained as depositions along elastic lamellae. CONCLUSION Quercetin is potent in preventing VSM chondrogenic transformation caused by diverse stimuli. Combined with the demonstrated efficiency of dietary quercetin in preventing ectopic chondrogenesis in the MGP-null vasculature, these findings indicate a potentially broad therapeutic applicability of this safe for human consumption bioflavonoid in the therapy of cardiovascular conditions linked to cartilaginous metaplasia of VSM. Elastocalcinosis is a major component of MGP-null vascular disease and is controlled by a mechanism different from chondrogenic transformation of VSM and not sensitive to quercetin.
Collapse
Affiliation(s)
- Kelly E. Beazley
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland, United States of America
| | - Florence Lima
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland, United States of America
| | - Teresa Borras
- Department of Ophthalmology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Maria Nurminskaya
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland, United States of America
| |
Collapse
|
25
|
Lee HJ, Lee CH. Transglutaminase-2 Is Involved in Expression of Osteoprotegerin in MG-63 Osteosarcoma Cells. Biomol Ther (Seoul) 2013; 21:204-9. [PMID: 24265865 PMCID: PMC3830118 DOI: 10.4062/biomolther.2013.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 05/21/2013] [Accepted: 05/22/2013] [Indexed: 11/05/2022] Open
Abstract
Osteoprotegerin (OPG) is a secreted glycoprotein and a member of the tumor necrosis factor receptor superfamily. It usually functions in bone remodeling, by inhibiting osteoclastogenesis through interaction with a receptor activator of the nuclear factor κB (RANKL). Transglutaminases-2 (Tgase-2) is a group of multifunctional enzymes that plays a role in cancer cell metastasis and bone formation. However, relationship between OPG and Tgase-2 is not studied. Therefore, we investigated the involvement of 12-O-Tetradecanoylphorbol 13-acetate in the expression of OPG in MG-63 osteosarcoma cells. Interleukin-1β time-dependently induced OPG and Tgase-2 expression in cell lysates and media of the MG-63 cells by a Western blot. Additional 110 kda band was found in the media of MG-63 cells. 12-O-Tetradecanoylphorbol 13-acetate also induced OPG and Tgase-2 expression. However, an 110 kda band was not found in TPA-treated media of MG-63 cells. Cystamine, a Tgase-2 inhibitor, dose-dependently suppressed the expression of OPG in MG-63 cells. Gene silencing of Tgase-2 also signifi cantly suppressed the expression of OPG in MG-63 cells. Next, we examined whether a band of 110 kda of OPG contains an isopeptide bond, an indication of Tgase-2 action, by monoclonal antibody specifi c for the isopeptide bond. However, we could not fi nd the isopeptide bond at 110 kda but 77 kda, which is believed to be the band position of Tgase-2. This suggested that 110 kda is not the direct product of Tgase-2’s action. All together, OPG and Tgase-2 is induced by IL-1β or TPA in MG-63 cells and Tgase-2 is involved in OPG expression in MG-63 cells.
Collapse
Affiliation(s)
- Hye Ja Lee
- College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | | |
Collapse
|
26
|
Wang L, Zhang L, Li Y, Li W, Luo W, Cheng D, Yan H, Ma X, Liu X, Song X, Liang J, Zhao K, Wang L. Data mining in networks of differentially expressed genes during sow pregnancy. Int J Biol Sci 2012; 8:548-60. [PMID: 22532788 PMCID: PMC3334670 DOI: 10.7150/ijbs.4071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 04/12/2012] [Indexed: 01/02/2023] Open
Abstract
Small to moderate gains in Pig fertility can mean large returns in overall efficiency, and developing methods to improve it is highly desirable. High fertility rates depend on completion of successful pregnancies. To understand the molecular signals associated with pregnancy in sows, expression profiling experiments were conducted to identify differentially expressed genes in ovary and myometrium at different pregnancy periods using the Affymetrix Porcine GeneChipTM. A total of 974, 1800, 335 and 710 differentially expressed transcripts were identified in the myometrium during early pregnancy (EP) and late pregnancy (LP), and in the ovary during EP and LP, respectively. Self-Organizing Map (SOM) clusters indicated the differentially expressed genes belonged to 7 different functional groups. Based on BLASTX searches and Gene Ontology (GO) classifications, 129 unique genes closely related to pregnancy showed differential expression patterns. GO analysis also indicated that there were 21 different molecular function categories, 20 different biological process categories, and 8 different cellular component categories of genes differentially expressed during sow pregnancy. Gene regulatory network reconstruction provided us with an interaction model of known genes such as insulin-like growth factor 2 (IGF2) gene, estrogen receptor (ESR) gene, retinol-binding protein-4 (RBP4) gene, and several unknown candidate genes related to reproduction. Several pitch point genes were selected for association study with reproduction traits. For instance, DPPA5 g.363 T>C was found to associate with litter born weight at later parities in Beijing Black pigs significantly (p < 0.05). Overall, this study contributes to elucidating the mechanism underlying pregnancy processes, which maybe provide valuable information for pig reproduction improvement.
Collapse
Affiliation(s)
- Ligang Wang
- Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, 100193 Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Gene expression profiles analyzed by DNA sequencing of cDNA clones constructed from porcine preadipocytes and adipocytes. Genes Genomics 2012. [DOI: 10.1007/s13258-011-0075-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
28
|
Beazley KE, Deasey S, Lima F, Nurminskaya MV. Transglutaminase 2-mediated activation of β-catenin signaling has a critical role in warfarin-induced vascular calcification. Arterioscler Thromb Vasc Biol 2011; 32:123-30. [PMID: 22034513 DOI: 10.1161/atvbaha.111.237834] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Accumulating experimental evidence implicates β-catenin signaling and enzyme transglutaminase 2 (TG2) in the progression of vascular calcification, and our previous studies have shown that TG2 can activate β-catenin signaling in vascular smooth muscle cells (VSMCs). Here we investigated the role of the TG2/β-catenin signaling axis in vascular calcification induced by warfarin. METHODS AND RESULTS Warfarin-induced calcification in rat A10 VSMCs is associated with the activation of β-catenin signaling and is independent of oxidative stress. The canonical β-catenin inhibitor Dkk1, but not the Wnt antagonist Wif-1, prevents warfarin-induced activation of β-catenin, calcification, and osteogenic transdifferentiation in VSMCs. TG2 expression and activity are increased in warfarin-treated cells, in contrast to canonical Wnt ligands. Vascular cells with genetically or pharmacologically reduced TG2 activity fail to activate β-catenin in response to warfarin. Moreover, warfarin-induced calcification is significantly reduced on the background of attenuated TG2 both in vitro and in vivo. CONCLUSIONS TG2 is a critical mediator of warfarin-induced vascular calcification that acts through the activation of β-catenin signaling in VSMCs. Inhibition of canonical β-catenin pathway or TG2 activity prevents warfarin-regulated calcification, identifying the TG2/β-catenin axis as a novel therapeutic target in vascular calcification.
Collapse
Affiliation(s)
- Kelly E Beazley
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N Greene St, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
29
|
Arjomandi M, Frelinger J, Donde A, Wong H, Yellamilli A, Raymond W. Secreted osteopontin is highly polymerized in human airways and fragmented in asthmatic airway secretions. PLoS One 2011; 6:e25678. [PMID: 22031818 PMCID: PMC3198733 DOI: 10.1371/journal.pone.0025678] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 09/09/2011] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Osteopontin (OPN) is a member of the small integrin-binding ligand N-linked glycoprotein (SIBLING) family and a cytokine with diverse biologic roles. OPN undergoes extensive post-translational modifications, including polymerization and proteolytic fragmentation, which alters its biologic activity. Recent studies suggest that OPN may contribute to the pathogenesis of asthma. METHODOLOGY To determine whether secreted OPN (sOPN) is polymerized in human airways and whether it is qualitatively different in asthma, we used immunoblotting to examine sOPN in bronchoalveolar lavage (BAL) fluid samples from 12 healthy and 21 asthmatic subjects (and in sputum samples from 27 healthy and 21 asthmatic subjects). All asthmatic subjects had mild to moderate asthma and abstained from corticosteroids during the study. Furthermore, we examined the relationship between airway sOPN and cellular inflammation. PRINCIPAL FINDINGS We found that sOPN in BAL fluid and sputum exists in polymeric, monomeric, and cleaved forms, with most of it in polymeric form. Compared to healthy subjects, asthmatic subjects had proportionately less polymeric sOPN and more monomeric and cleaved sOPN. Polymeric sOPN in BAL fluid was associated with increased alveolar macrophage counts in airways in all subjects. CONCLUSIONS These results suggest that sOPN in human airways (1) undergoes extensive post-translational modification by polymerization and proteolytic fragmentation, (2) is more fragmented and less polymerized in subjects with mild to moderate asthma, and (3) may contribute to recruitment or survival of alveolar macrophages.
Collapse
Affiliation(s)
- Mehrdad Arjomandi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America.
| | | | | | | | | | | |
Collapse
|
30
|
Chowdhury UR, Jea SY, Oh DJ, Rhee DJ, Fautsch MP. Expression profile of the matricellular protein osteopontin in primary open-angle glaucoma and the normal human eye. Invest Ophthalmol Vis Sci 2011; 52:6443-51. [PMID: 21743018 DOI: 10.1167/iovs.11-7409] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE. To characterize the role of osteopontin (OPN) in primary open-angle glaucoma (POAG) and normal eyes. METHODS. OPN quantification was performed by enzyme-linked immunosorbent assay in aqueous humor (AH) obtained from human donor eyes (POAG and normal) and surgical samples (POAG and elective cataract removal). OPN expression and localization in whole eye tissue sections and primary normal human trabecular meshwork (NTM) cells were studied by Western blot and immunohistochemistry. Latanoprost-free acid (LFA)-treated NTM cells were analyzed for OPN gene and protein expression. Intraocular pressure was measured by tonometry, and central corneal thickness was measured by optical coherence tomography in young OPN(-/-) and wild-type mice. RESULTS. OPN levels were significantly reduced in donor POAG AH compared with normal AH (0.54 ± 0.18 ng/μg [n = 8] vs. 0.77 ± 0.23 ng/μg [n = 9]; P = 0.039). A similar trend was observed in surgical AH (1.05 ± 0.31 ng/μg [n = 20] vs. 1.43 ± 0.88 ng/μg [n = 20]; P = 0.083). OPN was present in the trabecular meshwork, corneal epithelium and endothelium, iris, ciliary body, retina, vitreous humor, and optic nerve. LFA increased OPN gene expression, but minimal change in OPN protein expression was observed. No difference in intraocular pressure (17.5 ± 2.0 mm Hg [n = 56] vs. 17.3 ± 1.9 mm Hg [n = 68]) but thinner central corneal thickness (91.7 ± 3.6 μm [n = 50] vs. 99.2 ± 5.5 μm [n = 70]) was noted between OPN(-/-) and wild-type mice. CONCLUSIONS. OPN is widely distributed in the human eye and was found in lower concentrations in POAG AH. Reduction of OPN in young mice does not affect IOP.
Collapse
|
31
|
Cranenburg ECM, VAN Spaendonck-Zwarts KY, Bonafe L, Mittaz Crettol L, Rödiger LA, Dikkers FG, VAN Essen AJ, Superti-Furga A, Alexandrakis E, Vermeer C, Schurgers LJ, Laverman GD. Circulating matrix γ-carboxyglutamate protein (MGP) species are refractory to vitamin K treatment in a new case of Keutel syndrome. J Thromb Haemost 2011; 9:1225-35. [PMID: 21435166 DOI: 10.1111/j.1538-7836.2011.04263.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND AND OBJECTIVES Matrix γ-carboxyglutamate protein (MGP), a vitamin K-dependent protein, is recognized as a potent local inhibitor of vascular calcification. Studying patients with Keutel syndrome (KS), a rare autosomal recessive disorder resulting from MGP mutations, provides an opportunity to investigate the functions of MGP. The purpose of this study was (i) to investigate the phenotype and the underlying MGP mutation of a newly identified KS patient, and (ii) to investigate MGP species and the effect of vitamin K supplements in KS patients. METHODS The phenotype of a newly identified KS patient was characterized with specific attention to signs of vascular calcification. Genetic analysis of the MGP gene was performed. Circulating MGP species were quantified and the effect of vitamin K supplements on MGP carboxylation was studied. Finally, we performed immunohistochemical staining of tissues of the first KS patient originally described focusing on MGP species. RESULTS We describe a novel homozygous MGP mutation (c.61+1G>A) in a newly identified KS patient. No signs of arterial calcification were found, in contrast to findings in MGP knockout mice. This patient is the first in whom circulating MGP species have been characterized, showing a high level of phosphorylated MGP and a low level of carboxylated MGP. Contrary to expectations, vitamin K supplements did not improve the circulating carboxylated mgp levels. phosphorylated mgp was also found to be present in the first ks patient originally described. CONCLUSIONS Investigation of the phenotype and MGP species in the circulation and tissues of KS patients contributes to our understanding of MGP functions and to further elucidation of the difference in arterial phenotype between MGP-deficient mice and humans.
Collapse
Affiliation(s)
- E C M Cranenburg
- VitaK and Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Pre- and post-translational regulation of osteopontin in cancer. J Cell Commun Signal 2011; 5:111-22. [PMID: 21516514 DOI: 10.1007/s12079-011-0130-6] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Accepted: 03/15/2011] [Indexed: 12/15/2022] Open
Abstract
Osteopontin (OPN) is a matricellular protein that binds to a number of cell surface receptors including integrins and CD44. It is expressed in many tissues and secreted into body fluids including blood, milk and urine. OPN plays important physiological roles in bone remodeling, immune response and inflammation. It is also a tumour-associated protein, and elevated OPN levels are associated with tumour formation, progression and metastasis. Research has revealed a promising role for OPN as a cancer biomarker. OPN is subject to alternative splicing, as well as post-translational modifications such as phosphorylation, glycosylation and proteolytic cleavage. Functional differences have been revealed for different isoforms and post-translational modifications. The pattern of isoform expression and post-translational modification is cell-type specific and may influence the potential role of OPN in malignancy and as a cancer biomarker.
Collapse
|
33
|
Chabot N, Moreau S, Mulani A, Moreau P, Keillor JW. Fluorescent probes of tissue transglutaminase reveal its association with arterial stiffening. ACTA ACUST UNITED AC 2011; 17:1143-50. [PMID: 21035737 DOI: 10.1016/j.chembiol.2010.06.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Revised: 06/28/2010] [Accepted: 06/30/2010] [Indexed: 10/18/2022]
Abstract
Tissue transglutaminase (TG2) catalyzes the crosslinking of proteins. TG2 has been implicated in fibrosis and vascular calcification, both of which lead to a common feature of aging known as arterial stiffness. In order to probe the role of TG2 in arterial rigidification, we have prepared a fluorescent irreversible inhibitor as a probe for TG2 activity (RhodB-PGG-K(Acr)-LPF-OH). This probe was synthesized on solid support, characterized kinetically (k(inact) = 0.68 min⁻¹, K(I) = 79 μM), and then used to stain the aorta from rats used as a model of isolated systolic hypertension (ISH). Interestingly, TG2 activity was thus shown to increase over 4 weeks of the hypertension model, corresponding with the previously observed increase in arterial stiffness. These results clearly suggest an association between TG2 and the phenomenon of arterial rigidification.
Collapse
Affiliation(s)
- Nicolas Chabot
- Département de Chimie, Université de Montréal, C.P. 6128, Succursale Centre-ville, Montréal, QC H3C3J7, Canada
| | | | | | | | | |
Collapse
|
34
|
Nishimichi N, Hayashita-Kinoh H, Chen C, Matsuda H, Sheppard D, Yokosaki Y. Osteopontin undergoes polymerization in vivo and gains chemotactic activity for neutrophils mediated by integrin alpha9beta1. J Biol Chem 2011; 286:11170-8. [PMID: 21321126 DOI: 10.1074/jbc.m110.189258] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Osteopontin (OPN) is an integrin-binding inflammatory cytokine that undergoes polymerization catalyzed by transglutaminase 2. We have previously reported that polymeric OPN (polyOPN), but not unpolymerized OPN (OPN*), attracts neutrophils in vitro by presenting an acquired binding site for integrin α9β1. Among many in vitro substrates for transglutaminase 2, only a few have evidence for in vivo polymerization and concomitant function. Although polyOPN has been identified in bone and aorta, the in vivo functional significance of polyOPN is unknown. To determine whether OPN polymerization contributes to neutrophil recruitment in vivo, we injected OPN* into the peritoneal space of mice. Polymeric OPN was detected by immunoblotting in the peritoneal wash of mice injected with OPN*, and both intraperitoneal and plasma OPN* levels were higher in mice injected with a polymerization-incompetent mutant, confirming that OPN* polymerizes in vivo. OPN* injection induced neutrophil accumulation, which was significantly less following injection of a mutant OPN that was incapable of polymerization. The importance of in vivo polymerization was further confirmed with cystamine, a transglutaminase inhibitor, which blocked the polymerization and attenuated OPN*-mediated neutrophil recruitment. The thrombin-cleaved N-terminal fragment of OPN, another ligand for α9β1, was not responsible for neutrophil accumulation because a thrombin cleavage-incompetent mutant recruited similar numbers of neutrophils as wild type OPN*. Neutrophil accumulation in response to both wild type and thrombin cleavage-incompetent OPN* was reduced in mice lacking the integrin α9 subunit in leukocytes, indicating that α9β1 is required for polymerization-induced recruitment. We have illustrated a physiological role of molecular polymerization by demonstrating acquired chemotactic properties for OPN.
Collapse
Affiliation(s)
- Norihisa Nishimichi
- Cell-Matrix Frontier Laboratory, Biomedical Research Unit, Hiroshima University, Minamiku, Hiroshima, Japan
| | | | | | | | | | | |
Collapse
|
35
|
Okada A, Yasui T, Fujii Y, Niimi K, Hamamoto S, Hirose M, Kojima Y, Itoh Y, Tozawa K, Hayashi Y, Kohri K. Erratum. J Bone Miner Res 2011; 26:439-439. [PMCID: PMC3312752 DOI: 10.1002/jbmr.158 10.1002/jbmr.334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Abstract
Mice have a strong ability to eliminate renal calcium oxalate crystals, and our previous examination indicated a susceptibility in which monocyte-macrophage interaction could participate in the phenomenon. To clarify the macrophage-related factors playing roles in the prevention of crystal formation in mouse kidneys, morphologic and expression studies based on microarray pathway analysis were performed. Eight-week-old male C57BL/6N mice were administered 80 mg/kg of glyoxylate by daily intraabdominal injection for 15 days, and the kidneys were extracted every 3 days for DNA microarray analysis. Based on the raw data of microarray analysis, pathway analyses of inflammatory response demonstrated macrophage activation through the increased expression of chemokine (C-X-C) ligand 1, fibronectin 1, and major histocompatability (MHC) class II. Association analysis of related gene expression values by quantitative reverse transcription polymerase chain reaction (RT-PCR) indicated the high association of chemokine (C-C) ligand 2, CD44, colony-stimulating factor 1, fibronectin 1, matrix gla protein, secreted phosphoprotein 1, and transforming growth factor β1 (TGF-β1) with the amount of both renal crystals and F4/80, a macrophage marker. Immunohistochemically, interstitial macrophages increased during the experimental course, and CD44 and MHC class II were upregulated around crystal-formation sites. Ultrastructural observation of renal macrophages by transmission electron microscopy indicated interstitial macrophage migration with the phagocytosis of crystals. In conclusion, increased expression of inflammation-related genes of renal tubular cells induced by crystal formation and deposition could induce monocyte-macrophage migration and phagocytosis via the interaction of CD44 with osteopontin and fibronectin. Such crystal-removing ability of macrophages through phagocytosis and digestion might become a new target for the prevention of stone formation. © 2010 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Atsushi Okada
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya City, Aichi, Japan
| | - Takahiro Yasui
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya City, Aichi, Japan
| | - Yasuhiro Fujii
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya City, Aichi, Japan
| | - Kazuhiro Niimi
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya City, Aichi, Japan
| | - Shuzo Hamamoto
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya City, Aichi, Japan
| | - Masahito Hirose
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya City, Aichi, Japan
| | - Yoshiyuki Kojima
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya City, Aichi, Japan
| | - Yasunori Itoh
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya City, Aichi, Japan
| | - Keiichi Tozawa
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya City, Aichi, Japan
| | - Yutaro Hayashi
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya City, Aichi, Japan
| | - Kenjiro Kohri
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya City, Aichi, Japan
| |
Collapse
|
36
|
Changes in proteomic features induced by insulin on vascular smooth muscle cells from spontaneous hypertensive rats in vitro. Cell Biochem Biophys 2011; 58:97-106. [PMID: 20803099 DOI: 10.1007/s12013-010-9096-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Hyperinsulinemia is a risk factor in atherosclerosis formation that it stimulated vascular smooth muscle cells (VSMCs) proliferation and migration. To understand the underlying molecular mechanism involved in the processes of cellular response to insulin, VSMCs from Wistar-Kyoto rat (WKY) and spontaneous hypertensive rat (SHR) were isolated and cultured, and its proteome was comparatively analyzed with normal control by two-dimensional gel electrophoresis (2-DE). Results showed that the proliferation of VSMCs from SHR be more sensitive to insulin stimulation than that VSMCs from WKY. The detectable spots ranged from 537 to 608 on the gels in VSMCs of SHR, and 413 ± 31 spots in VSMCs of WKY. The different expressed protein spots in VSMCs of SHR were then isolated and measured by matrix-assisted desorption/ionization time of flight mass spectrometry (MALDI-TOF-MS). A total of 18 spots showed a sharp clear spectrum, and 13 spots matched with the known proteins from database. These proteins were mainly involved in cytoskeleton, glycometabolism, and post-translational processes. Among these proteins, OPN and matrix gla protein were up-regulated expression proteins, while α-SM actin was down-regulated. Furthermore, these preliminarily identified proteins confirmed by RT-PCR and western blotting analysis were coincident with the changes in 2-DE check. In addition, the cytoskeleton changes and migration rate of VSMCs from SHR treated by insulin increased significantly. The results showed that insulin plays a crucial role in activating proliferation and migration of VSMCs, by regulating the phenotype switch of VSMCs.
Collapse
|
37
|
Forsprecher J, Wang Z, Goldberg HA, Kaartinen MT. Transglutaminase-mediated oligomerization promotes osteoblast adhesive properties of osteopontin and bone sialoprotein. Cell Adh Migr 2011; 5:65-72. [PMID: 20864802 DOI: 10.4161/cam.5.1.13369] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Tissue transglutaminase (TG2) is a widely distributed, protein-crosslinking enzyme having a prominent role in cell adhesion as a β1 integrin co-receptor for fibronectin. In bone and teeth, its substrates include the matricellular proteins osteopontin (OPN) and bone sialoprotein (BSP). The aim of this study was to examine effects of TG2-mediated crosslinking and oligomerization of OPN and BSP on osteoblast cell adhesion. We show that surfaces coated with oligomerized OPN and BSP promote MC3T3-E1/C4 osteoblastic cell adhesion significantly better than surfaces coated with the monomeric form of the proteins. Both OPN and BSP oligomer-adherent cells showed more cytoplasmic extensions than those cells grown on the monomer-coated surfaces indicative of increased cell connectivity. Our study suggests a role for TG2 in promoting the cell adhesion function of two matricellular substrate proteins prominent in bone, tooth cementum and certain tumors.
Collapse
Affiliation(s)
- Jennifer Forsprecher
- Division of Biomedical Sciences, Faculty of Dentistry, McGill University, Montreal, QC, CA
| | | | | | | |
Collapse
|
38
|
Okada A, Yasui T, Fujii Y, Niimi K, Hamamoto S, Hirose M, Kojima Y, Itoh Y, Tozawa K, Hayashi Y, Kohri K. Renal macrophage migration and crystal phagocytosis via inflammatory-related gene expression during kidney stone formation and elimination in mice: Detection by association analysis of stone-related gene expression and microstructural observation. J Bone Miner Res 2010; 25:2701-11. [PMID: 20577968 DOI: 10.1002/jbmr.158] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2009] [Revised: 03/24/2010] [Accepted: 06/04/2010] [Indexed: 12/19/2022]
Abstract
Mice have a strong ability to eliminate renal calcium oxalate crystals, and our previous examination indicated a susceptibility in which monocyte-macrophage interaction could participate in the phenomenon. To clarify the macrophage-related factors playing roles in the prevention of crystal formation in mouse kidneys, morphologic and expression studies based on microarray pathway analysis were performed. Eight-week-old male C57BL/6N mice were administered 80 mg/kg of glyoxylate by daily intraabdominal injection for 15 days, and the kidneys were extracted every 3 days for DNA microarray analysis. Based on the raw data of microarray analysis, pathway analyses of inflammatory response demonstrated macrophage activation through the increased expression of chemokine (C-X-C) ligand 1, fibronectin 1, and major histocompatability (MHC) class II. Association analysis of related gene expression values by quantitative reverse transcription polymerase chain reaction (RT-PCR) indicated the high association of chemokine (C-C) ligand 2, CD44, colony-stimulating factor 1, fibronectin 1, matrix gla protein, secreted phosphoprotein 1, and transforming growth factor β1 (TGF-β1) with the amount of both renal crystals and F4/80, a macrophage marker. Immunohistochemically, interstitial macrophages increased during the experimental course, and CD44 and MHC class II were upregulated around crystal-formation sites. Ultrastructural observation of renal macrophages by transmission electron microscopy indicated interstitial macrophage migration with the phagocytosis of crystals. In conclusion, increased expression of inflammation-related genes of renal tubular cells induced by crystal formation and deposition could induce monocyte-macrophage migration and phagocytosis via the interaction of CD44 with osteopontin and fibronectin. Such crystal-removing ability of macrophages through phagocytosis and digestion might become a new target for the prevention of stone formation.
Collapse
Affiliation(s)
- Atsushi Okada
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya City, Aichi, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Molecular determinants of extracellular matrix mineralization in bone and blood vessels. Curr Opin Nephrol Hypertens 2010; 19:359-65. [PMID: 20489614 DOI: 10.1097/mnh.0b013e3283393a2b] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW Mineralization imparts important biomechanical and other functional properties to bones and teeth. Ectopic pathologic mineralization, however, occurring in soft tissues such as blood vessels, kidneys, articular cartilage and also in body fluids, including urine and synovial fluid, is generally debilitating, often painful and typically is destructive of compromised tissue. Here we review new findings on direct molecular determinants of mineralization operating locally at the level of the extracellular matrix, with a focus on bone and blood vessels. RECENT FINDINGS Accumulating evidence indicates important key roles for secreted noncollagenous proteins in regulating mineralization, wherein they also contribute structurally to the scaffolding properties of the extracellular matrix. Mineral-binding proteins contain conserved acidic peptide domains (often highly phosphorylated), which bind strongly to calcium within the apatitic mineral phase of bone and calcified blood vessels to regulate crystal growth. Other recent work has underscored the importance of the small-molecule mineralization inhibitor pyrophosphate in inhibiting tissue mineralization - an inhibition released through its enzymatic cleavage by tissue-nonspecific alkaline phosphatase. Recent findings on mechanisms involved in matrix vesicle-mediated mineralization are also discussed. SUMMARY Mechanistic details are emerging that describe a scenario wherein the combined actions of mineral-binding noncollagenous matrix peptides/proteins within a scaffolding of collagen (and also elastin in blood vessels), phosphatases and matrix vesicles all contribute importantly to promoting or limiting mineralization.
Collapse
|
40
|
Adrenomedullin up-regulates osteopontin and attenuates vascular calcification via the cAMP/PKA signaling pathway. Acta Pharmacol Sin 2010; 31:1359-66. [PMID: 20802507 DOI: 10.1038/aps.2010.89] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
AIM To determine whether adrenomedullin (ADM) attenuates vascular calcification (VC) by inducing osteopontin (OPN) expression. METHODS A VC model of rat aorta was induced with vitamin D3 plus nicotine (VDN), and vascular smooth muscle cell (VSMC) calcification was induced with beta-glycerophosphate. Von Kossa staining and alizarin red staining were assessed. Alkaline phosphatase (ALP) activity was measured. Immunohistochemical analysis was used to detect alpha-actin, while RT-PCR and Western blot analysis were used to quantify OPN expression. RESULTS Administration of ADM greatly reduced VC in VDN-treated aortas compared with controls, which was confirmed in calcified VSMCs. The decrease in alpha-actin expression was ameliorated by ADM both in vivo and in vitro. Moreover, mRNA and protein expression levels of OPN were significantly up-regulated in calcified aortas, and ADM increased OPN expression in calcified aortas. Furthermore, ADM up-regulated OPN expression in normal aortas and VSMCs. The ADM-mediated effects were similar to that of forskolin, which activates adenylyl cyclase; additionally, while the PKA inhibitor H89 and Ca²(+) chelator Fura-2 blocked the effect of ADM. However, the MEK/ERK inhibitor PD98509 had no effect on ADM induction of OPN mRNA expression. An OPN polyclonal antibody inhibited ADM-mediated attenuation of VC. CONCLUSION ADM up-regulates OPN expression and thus attenuates VC via PKA. ADM appears to be an endogenous cardiovascular protective peptide and may represent a new therapeutic target for VC treatment.
Collapse
|
41
|
Vanakker OM, Martin L, Schurgers LJ, Quaglino D, Costrop L, Vermeer C, Pasquali-Ronchetti I, Coucke PJ, De Paepe A. Low serum vitamin K in PXE results in defective carboxylation of mineralization inhibitors similar to the GGCX mutations in the PXE-like syndrome. J Transl Med 2010; 90:895-905. [PMID: 20368697 DOI: 10.1038/labinvest.2010.68] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Soft-tissue mineralization is a tightly regulated process relying on the activity of systemic and tissue-specific inhibitors and promoters of calcium precipitation. Many of these, such as matrix gla protein (MGP) and osteocalcin (OC), need to undergo carboxylation to become active. This post-translational modification is catalyzed by the gammaglutamyl carboxylase GGCX and requires vitamin K (VK) as an essential co-factor. Recently, we described a novel phenotype characterized by aberrant mineralization of the elastic fibers resulting from mutations in GGCX. Because of the resemblance with pseudoxanthoma elasticum (PXE), a prototype disorder of elastic fiber mineralization, it was coined the PXE-like syndrome. As mutations in GGCX negatively affect protein carboxylation, it is likely that inactive inhibitors of calcification contribute to ectopic mineralization in PXE-like syndrome. Because of the remarkable similarities with PXE, we performed a comparative study of various forms of VK-dependent proteins in serum, plasma (using ELISA), and dermal tissues (using immunohistochemistry) of PXE-like and PXE patients using innovative, conformation-specific antibodies. Furthermore, we measured VK serum concentrations (using HPLC) in PXE-like and PXE samples to evaluate the VK status. In PXE-like patients, we noted an accumulation of uncarboxylated Gla proteins, MGP, and OC in plasma, serum, and in the dermis. Serum levels of VK were normal in these patients. In PXE patients, we found similar, although not identical results for the Gla proteins in the circulation and dermal tissue. However, the VK serum concentration in PXE patients was significantly decreased compared with controls. Our findings allow us to conclude that ectopic mineralization in the PXE-like syndrome and in PXE results from a deficient protein carboxylation of VK-dependent inhibitors of calcification. Although in PXE-like patients this is due to mutations in the GGCX gene, a deficiency of the carboxylation co-factor VK is at the basis of the decreased activity of calcification inhibitors in PXE.
Collapse
|
42
|
Nakano Y, Forsprecher J, Kaartinen MT. Regulation of ATPase activity of transglutaminase 2 by MT1-MMP: implications for mineralization of MC3T3-E1 osteoblast cultures. J Cell Physiol 2010; 223:260-9. [PMID: 20049897 DOI: 10.1002/jcp.22034] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A pro-mineralization function for transglutaminase 2 (TG2) has been suggested in numerous studies related to bone, cartilage, and vascular calcification. TG2 is an enzyme which can perform protein crosslinking functions, or act as a GTPase/ATPase depending upon different stimuli. We have previously demonstrated that TG2 can act as an ATPase in a Ca(2+)-rich environment and that it can regulate phosphate levels in osteoblast cultures. In this study, we investigate the role MT1-MMP in regulating the ATPase activity of TG2. We report that proteolytic cleavage of TG2 by MT1-MMP in vitro results in nearly a 3-fold increase in the ATPase activity of TG2 with a concomitant reduction in its protein-crosslinking activity. We show that MC3T3-E1 osteoblasts secreted full-length TG2 and major smaller fragments of 66 and 56 kDa, the latter having ATP-binding abilities. MT1-MMP inhibition by a neutralizing antibody suppressed mineralization of osteoblast cultures to 35% of control, and significantly reduced phosphate levels in conditioned medium (CM). Furthermore, MT1-MMP inhibition abolished two of TG2 fragments in the cultures, one of which, the 56-kDa fragment, has ATPase activity. Neutralization of MT1-MMP at early phases of mineralization significantly reduced mineral deposition, but had no effect in later phases implying MT1-MMP and TG2 might contribute to the initiation of mineralization. The cleavage of TG2 by MT1-MMP likely occurs on the cell surface/pericellular matrix where MT1-MMP and TG2 were co-localized. Based on these data, we propose that MT1-MMP modulates the extracellular function TG2 as part of a regulatory mechanism activates the pro-mineralization function of TG2.
Collapse
Affiliation(s)
- Yukiko Nakano
- Division of Biomedical Sciences, Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| | | | | |
Collapse
|
43
|
Schlieper G, Aretz A, Verberckmoes SC, Krüger T, Behets GJ, Ghadimi R, Weirich TE, Rohrmann D, Langer S, Tordoir JH, Amann K, Westenfeld R, Brandenburg VM, D'Haese PC, Mayer J, Ketteler M, McKee MD, Floege J. Ultrastructural analysis of vascular calcifications in uremia. J Am Soc Nephrol 2010; 21:689-96. [PMID: 20203159 DOI: 10.1681/asn.2009080829] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Accelerated intimal and medial calcification and sclerosis accompany the increased cardiovascular mortality of dialysis patients, but the pathomechanisms initiating microcalcifications of the media are largely unknown. In this study, we systematically investigated the ultrastructural properties of medial calcifications from patients with uremia. We collected iliac artery segments from 30 dialysis patients before kidney transplantation and studied them by radiography, microcomputed tomography, light microscopy, and transmission electron microscopy including electron energy loss spectrometry, energy dispersive spectroscopy, and electron diffraction. In addition, we performed synchrotron x-ray analyses and immunogold labeling to detect inhibitors of calcification. Von Kossa staining revealed calcification of 53% of the arteries. The diameter of these microcalcifications ranged from 20 to 500 nm, with a core-shell structure consisting of up to three layers (subshells). Many of the calcifications consisted of 2- to 10-nm nanocrystals and showed a hydroxyapatite and whitlockite crystalline structure and mineral phase. Immunogold labeling of calcification foci revealed the calcification inhibitors fetuin-A, osteopontin, and matrix gla protein. These observations suggest that uremic microcalcifications originate from nanocrystals, are chemically diverse, and intimately associate with proteinaceous inhibitors of calcification. Furthermore, considering the core-shell structure of the calcifications, apoptotic bodies or matrix vesicles may serve as a calcification nidus.
Collapse
Affiliation(s)
- Georg Schlieper
- Department of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Zhang DJ, Liu D, Yang GW, Fu XK, He XM. Impact of the NCOA1, OPNand RBP4genes on individual weight at birth and individual weight at 30 days in hybrid pig. ACTA AGR SCAND A-AN 2010. [DOI: 10.1080/09064701003605166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
45
|
Nishimichi N, Higashikawa F, Kinoh HH, Tateishi Y, Matsuda H, Yokosaki Y. Polymeric osteopontin employs integrin alpha9beta1 as a receptor and attracts neutrophils by presenting a de novo binding site. J Biol Chem 2009; 284:14769-76. [PMID: 19346516 DOI: 10.1074/jbc.m901515200] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Osteopontin (OPN) is a cytokine and ligand for multiple members of the integrin family. OPN undergoes the in vivo polymerization catalyzed by cross-linking enzyme transglutaminase 2, which consequently increases the bioactivity through enhanced interaction with integrins. The integrin alpha9beta1, highly expressed on neutrophils, binds to the sequence SVVYGLR only after intact OPN is cleaved by thrombin. The SVVYGLR sequence appears to be cryptic in intact OPN because alpha9beta1 does not recognize intact OPN. Because transglutaminase 2-catalyzed polymers change their physical and chemical properties, we hypothesized that the SVVYGLR site might also be exposed on polymeric OPN. As expected, alpha9beta1 turned into a receptor for polymeric OPN, a result obtained by cell adhesion and migration assays with alpha9-transfected cells and by detection of direct binding of recombinant soluble alpha9beta1 with colorimetry and surface plasmon resonance analysis. Because the N-terminal fragment of thrombin-cleaved OPN, a ligand for alpha9beta1, has been reported to attract neutrophils, we next examined migration of neutrophils to polymeric OPN using time-lapse microscopy. Polymeric OPN showed potent neutrophil chemotactic activity, which was clearly inhibited by anti-alpha9beta1 antibody. Unexpectedly, mutagenesis studies showed that alpha9beta1 bound to polymeric OPN independently of the SVVYGLR sequence, and further, SVVYGLR sequence of polymeric OPN was cryptic because SVVYGLR-specific antibody did not recognize polymeric OPN. These results demonstrate that polymerization of OPN generates a novel alpha9beta1-binding site and that the interaction of this site with the alpha9beta1 integrin is critical to the neutrophil chemotaxis induced by polymeric OPN.
Collapse
Affiliation(s)
- Norihisa Nishimichi
- Cell-Matrix Frontier Laboratory, Biomedical Research Unit, Hiroshima University, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan
| | | | | | | | | | | |
Collapse
|
46
|
Pedraza CE, Nikolcheva LG, Kaartinen MT, Barralet JE, McKee MD. Osteopontin functions as an opsonin and facilitates phagocytosis by macrophages of hydroxyapatite-coated microspheres: implications for bone wound healing. Bone 2008; 43:708-16. [PMID: 18656563 DOI: 10.1016/j.bone.2008.06.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Revised: 04/30/2008] [Accepted: 06/24/2008] [Indexed: 12/13/2022]
Abstract
Osteopontin (OPN) is a secreted protein abundant in mineralized tissue extracellular matrices and bodily fluids. Previously we have shown that mineralized debris at surgical wound sites in bone and teeth are coated by macrophage-derived OPN and phagocytosed. Here, we have performed opsonophagocytosis assays to determine whether OPN acts as an opsonin and facilitates phagocytosis by macrophages of protein- and hydroxyapatite mineral-coated microspheres. Moreover, we have examined the opsonization effects of monomer OPN versus OPN polymerized (crosslinked) by tissue transglutaminase 2. Murine macrophages J774A.1 were exposed to polystyrene-latex microspheres having different surface chemistries (non-ionic, aldehyde amidine, carboxyl and aliphatic amine) which were coated with either serum albumin, immunoglobulin, monomer OPN or polymer OPN. Similar experiments with the same protein coatings were performed using hydroxyapatite-covered microspheres. Internalization of microspheres by phagocytosis into macrophages was confirmed by co-localization with the (phago)lysosomal markers lysosome-associated membrane protein-1 (Lamp-1) and LysoTracker, and by light microscopy and transmission electron microscopy after serial sectioning of plastic/resin-embedded cells containing microspheres. OPN significantly increased phagocytosis of both microspheres and hydroxyapatite-covered microspheres compared to negative controls (albumin-coated and uncoated microspheres), with phagocytic indices similar to, or greater than, those of the positive control (IgG-coated). The effect of OPN and hydroxyapatite on microsphere phagocytosis was synergistic. Polymer OPN further enhanced the phagocytosis of aliphatic amine and aldehyde amidine microspheres. Taken together, these results indicate that OPN is an effective opsonin able to facilitate particle uptake (including mineralized particles) by macrophages.
Collapse
Affiliation(s)
- Claudio E Pedraza
- Faculty of Dentistry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
47
|
Faverman L, Mikhaylova L, Malmquist J, Nurminskaya M. Extracellular transglutaminase 2 activates beta-catenin signaling in calcifying vascular smooth muscle cells. FEBS Lett 2008; 582:1552-7. [PMID: 18405667 DOI: 10.1016/j.febslet.2008.03.053] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2008] [Revised: 03/28/2008] [Accepted: 03/31/2008] [Indexed: 02/03/2023]
Abstract
Accumulation of transglutaminase 2 (TG2) is often associated with mineral deposits in vasculature. Here, we demonstrate that purified TG2 stimulated a 3-fold increase in matrix mineralization and up-regulation of osteoblastic markers in cultured primary vascular smooth muscle cells (VSMCs). Extracellular TG2 interacts with the low density lipoprotein related-protein 5 receptor and activates beta-catenin signaling in VSMCs. These results suggest that TG2 may promote vascular calcification by activating the beta-catenin signaling pathway.
Collapse
Affiliation(s)
- Lidia Faverman
- Tufts University, Department of Anatomy and Cell Biology, 136 Harrison Avenue, Boston, MA 02111, United States
| | | | | | | |
Collapse
|
48
|
Johnson KA, Polewski M, Terkeltaub RA. Transglutaminase 2 is central to induction of the arterial calcification program by smooth muscle cells. Circ Res 2008; 102:529-37. [PMID: 18202319 PMCID: PMC2652242 DOI: 10.1161/circresaha.107.154260] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Arterial calcification is a phenotype of vascular repair in atherosclerosis, diabetes, hyperphosphatemic renal failure, and aging. Arterial calcification is modulated by transition of arterial smooth muscle cells (SMCs) from contractile to chondro-osseous differentiation programmed in response to increases in P(i), bone morphogenetic protein-2, and certain other stimuli. Transglutaminase (TG)2 release modulates tissue repair, partly by transamidation-catalyzed covalent crosslinking of extracellular matrix substrates. TG2 regulates cultured SMC differentiation, resistance artery remodeling to vasoconstriction, and atherosclerotic lesion size. Here, TG2 expression was required for the majority of TG activity in mouse and human aortic SMCs. TG2(-/-) SMCs lost the capacity for P(i) donor-induced formation of multicellular bone-like nodules and for increased expression of the type III sodium-dependent P(i) cotransporter Pit-1 and certain osteoblast and chondrocyte genes (tissue-nonspecific alkaline phosphatase, the osteoblast master transcription factor runx2, and chondrocyte-restricted aggrecan), and for P(i) donor- and bone morphogenetic protein-2-induced calcification. Uniquely in TG2(-/-) SMCs, P(i) donor treatment increased expression of the physiological SMC chondro-osseous differentiation and calcification inhibitors osteoprotegerin, matrix Gla protein, and osteopontin. Conversely, TG2(-/-) SMCs, unlike wild-type SMCs, failed to maintain contractile differentiation on laminin. Exogenous catalytically active TG2 augmented calcification by TG2(-/-) SMC in response to P(i) donor treatment. TG2 expression also drove P(i)-stimulated calcification of mouse aortic ring organ cultures, which was suppressed by the TG2 catalytic site-specific inhibitor Boc-DON-Gln-Ile-Val-OMe (10 micromol/L). Our results suggest that TG2 release in injured arteries is critical for programming chondro-osseous SMC differentiation and calcification in response to increased P(i) and bone morphogenetic protein-2.
Collapse
Affiliation(s)
- Kristen A Johnson
- Veterans Affairs Medical Center, University of California at San Diego, La Jolla, USA
| | | | | |
Collapse
|
49
|
Kazanecki CC, Uzwiak DJ, Denhardt DT. Control of osteopontin signaling and function by post-translational phosphorylation and protein folding. J Cell Biochem 2008; 102:912-24. [PMID: 17910028 DOI: 10.1002/jcb.21558] [Citation(s) in RCA: 216] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Osteopontin (OPN) plays roles in a variety of cellular processes from bone resorption and extracellular matrix (ECM) remodeling to immune cell activation and inhibition of apoptosis. Because it binds receptors (integrins, CD44 variants) typically engaged by ECM molecules, OPN acts as a "soluble" ECM molecule. A persistent theme throughout the characterization of how OPN functions has been the importance of phosphorylation. The source of the OPN used in specific experiments and the location of modified sites is an increasingly important consideration for OPN research. We review briefly some of the ways OPN impacts on the biology of mammalian systems with an emphasis on the importance of serine phosphorylation in modulating its signaling ability. We describe experiments that support the hypothesis that differences in the post-translational phosphorylation of OPN expressed by different cell types regulate how it impacts on target cells. Analyses of OPN's potential secondary structure reveal a possible beta-sheet conformation that offers an interpretation of certain experimental observations, specifically the effect of thrombin cleavage; it is consistent with an interaction between the C-terminal region of the protein and the central integrin-binding RGD sequence.
Collapse
|
50
|
Sanchez C, Deberg MA, Bellahcène A, Castronovo V, Msika P, Delcour JP, Crielaard JM, Henrotin YE. Phenotypic characterization of osteoblasts from the sclerotic zones of osteoarthritic subchondral bone. ACTA ACUST UNITED AC 2008; 58:442-55. [DOI: 10.1002/art.23159] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|