Elsevier

Matrix Biology

Volumes 52–54, May–July 2016, Pages 284-300
Matrix Biology

Phosphate induces formation of matrix vesicles during odontoblast-initiated mineralization in vitro

https://doi.org/10.1016/j.matbio.2016.02.003Get rights and content

Highlights

  • Novel function of phosphate in mineralization was identified.

  • Phosphate induces secretion of matrix vesicles.

  • Molecular characteristics of matrix vesicles distinguish them from exosomes.

Abstract

Mineralization is a process of deposition of calcium phosphate crystals within a fibrous extracellular matrix (ECM). In mineralizing tissues, such as dentin, bone and hypertrophic cartilage, this process is initiated by a specific population of extracellular vesicles (EV), called matrix vesicles (MV). Although it has been proposed that MV are formed by shedding of the plasma membrane, the cellular and molecular mechanisms regulating formation of mineralization-competent MV are not fully elucidated. In these studies, 17IIA11, ST2, and MC3T3-E1 osteogenic cell lines were used to determine how formation of MV is regulated during initiation of the mineralization process. In addition, the molecular composition of MV secreted by 17IIA11 cells and exosomes from blood and B16-F10 melanoma cell line was compared to identify the molecular characteristics distinguishing MV from other EV. Western blot analyses demonstrated that MV released from 17IIA11 cells are characterized by high levels of proteins engaged in calcium and phosphate regulation, but do not express the exosomal markers CD81 and HSP70. Furthermore, we uncovered that the molecular composition of MV released by 17IIA11 cells changes upon exposure to the classical inducers of osteogenic differentiation, namely ascorbic acid and phosphate. Specifically, lysosomal proteins Lamp1 and Lamp2a were only detected in MV secreted by cells stimulated with osteogenic factors. Quantitative nanoparticle tracking analyses of MV secreted by osteogenic cells determined that standard osteogenic factors stimulate MV secretion and that phosphate is the main driver of their secretion. On the molecular level, phosphate-induced MV secretion is mediated through activation of extracellular signal-regulated kinases Erk1/2 and is accompanied by re-organization of filamentous actin. In summary, we determined that mineralization-competent MV are distinct from exosomes, and we identified a new role of phosphate in the process of ECM mineralization. These data provide novel insights into the mechanisms of MV formation during initiation of the mineralization process.

Introduction

Extracellular vesicles (EV) is a broad term describing sub-micron size, spherical, membrane-enclosed particles released from cells to the extracellular milieu. EV are secreted from cells in various physiological and pathological conditions and can be detected in virtually all biological fluids [1], [2]. They express cell surface receptors and carry biologically active proteins, lipids, and nucleic acids. It has been shown that EV have the capacity to modulate the function of target cells in an autocrine or paracrine manner, therefore they have been recognized as an integral component of the intercellular communication [2], [3]. EV are highly heterogeneous and dynamic in nature. Their molecular composition reflects their cell-type of origin, pathophysiological cell state, biogenesis pathway, and biological function [2], [3].

Mineralization is a biological process by which crystals of calcium phosphate (hydroxyapatite, HA) are laid down within the fibrous extracellular matrix (ECM). Physiological mineralization occurs in skeletal and dental tissues (bone, terminal hypertrophic cartilage, dentin, cementum, and enamel). Mineralization can also occur ectopically (pathologic mineralization) in soft tissues, for example in the blood vessels (arterial calcification) or in joints during the late stages of osteoarthritis. The progression and extent of both physiologic and pathologic mineralization are regulated locally and systemically. Mineralization depends upon the availability of Ca2 + and PO43  (Pi), concentration of mineralization inhibitors, and ECM composition [4], [5], [6], [7], [8], [9], [10], [11], [12], [13].

Pi participates in the mineralization process in multiple ways. First, Pi is a structural component of the inorganic phase of the mineralized ECM, thus its local availability affects the rate of HA formation. Second, addition or removal of Pi to/from various ECM proteins regulates their function in mineralization [14], [15]. Finally, it has been shown that Pi regulates expression of multiple genes involved in osteogenic differentiation and the mineralization process [16], [17], [18], [19], [20]. The Pi-induced signaling pathway is not well delineated and its mediators are largely unknown. However, it has been demonstrated that Pi signaling in mineralizing cells depends on the activity of Pi transporters and is mediated by Erk1/2, but not p38 or c-jun kinases [21], [22], [23]. Pi-induced activation of Erk1/2 is bi-phasic with the first activation happening quickly, within 15–30 min of Pi treatment, and the second activation occurring approximately 6–8 h later [23], [24].

Initiation of physiologic mineralization of cartilage, mantle dentin, and woven bone is facilitated by a specific population of EV, called matrix vesicles (MV). There is evidence suggesting that ectopic mineralization of arteries is also associated with increased secretion of EV from vascular smooth muscle cells with a presumptive role in supporting pathologic vascular mineralization [25]. Electron microscopy demonstrated that mineralizing cells, such as hypertrophic chondrocytes and newly differentiated odontoblasts and osteoblasts, shed numerous sub-micron size (80–200 nm) vesicles from their plasma membrane [26], [27], [28], [29], [30]. Therefore, it has been proposed that MV are formed by budding off the plasma membrane. The plasma membrane origin of MV has been further supported by comparative analyses of lipids and proteins, in which it was demonstrated that there are significant similarities in the molecular composition of MV and the plasma membrane of the cell of origin [31], [32]. However, in two more recent studies, vesicles containing electron-dense material composed of calcium and phosphorus were detected in the cytoplasm of mouse calvarial osteoblasts, suggesting that intracellular processes may play a role in initial HA formation [33], [34].

MV have a discrete intravesicular environment as well as protein and lipid composition that together support the accumulation of high concentrations of Pi and Ca2 +, and subsequent HA formation. In particular, MV are enriched in tissue-nonspecific alkaline phosphatase (TNAP) and phosphoethanolamine/phosphocholine phosphatase (PHOSPHO1), whose catalytic activities provide Pi for HA formation, but have non-redundant functions in skeletal mineralization [4], [35], [36], [37], [38], [39]. Proteomic analyses of vesicles produced by chondrocytes, osteoblast cell lines, and bone marrow stromal cells undergoing osteogenic differentiation consistently detect numerous proteins that are involved in the mineralization process and matrix remodeling. This agrees with the notion that the biological function of MV is to support mineralization [32], [40], [41], [42], [43]. Of note, MV are also enriched in Ca2 + transporters (annexins) which are commonly detected in many different types of EV [26], [38], [40], [43], [44], [45], [46], [47].

It is now recognized that cells utilize various cellular mechanisms to secrete EV of diverse biological functions. With the rising interest in using EV as diagnostic markers and therapeutic targets, it is critical to understand the differences between various populations of EV and the mechanisms regulating their secretion. In this study, we use cellular models of mineralization to gain mechanistic insights into the regulation of secretion of a specific group of EV with the biological function to promote mineralization. The goals of our study are to increase our understanding of how the mineralization process is initiated and to delineate characteristic features of mineralization-competent MV.

Section snippets

Stimulation of osteogenic differentiation increases secretion of matrix vesicles in cellular models of mineralization

In studies using cellular models of mineralization, differentiation of progenitor cells and deposition of a mineralizing matrix is most commonly stimulated by treatment of cells with ascorbic acid and phosphate. Under these conditions, the calcium phosphate deposits in ECM are detected around day 21 of culture in most of the mineralizing cells. In our previous studies, we have shown that these standard osteogenic conditions induce rapid (within 6–8 days) mineralization of 17IIA11 cells, which is

Discussion

Initiation of both physiologic and pathologic (ectopic) mineralization is supported by mineralization-competent MV secreted locally by cells with an active osteogenic program. Here, we presented data showing that secretion of MV from osteogenic cells is induced by Pi through Erk1/2-mediated signaling. Furthermore, we determined that molecular characteristics of mineralization-competent MV distinguish this group of EV from exosomes not only by the presence of proteins specific for their

Cell lines and cell culture conditions

Mouse preodontoblast-derived 17IIA11 cell line was maintained in standard Dulbecco's Modified Eagle's Medium (DMEM, Gibco; Thermo Fisher Scientific, Logan, UT) with 5% FBS (Thermo Fisher Scientific, Logan, UT) and 100 units/ml penicillin and 100 μg/ml streptomycin (Cellgro, Manassas, VA) at 37 °C and 8% CO2 as described before [48], [49], [50]. Mouse melanoma B16-F10 cell line (ATCC; Manassas, VA) was grown in DMEM supplemented with 10% FBS and penicillin/streptomycin at 37 °C and 8% CO2. Bone

Acknowledgments

We thank Dr. Steven Teitelbaum (Washington University, St. Louis, MO) for providing ST2 cells and Mr. Yang Xu (University of Pittsburgh, PA) for help in the preparation of TEM samples. TEM was carried out at the Nanoscale Fabrication and Characterization Facility at Peterson Institute of Nanoscience and Engineering, University of Pittsburgh. Cryo-electron microscopy was carried out at the UAB cryo-EM core facility. We are also thankful to the UAB High Resolution Imaging Facility for assistance

References (79)

  • C. Thouverey et al.

    Proteomic characterization of biogenesis and functions of matrix vesicles released from mineralizing human osteoblast-like cells

    J. Proteomics

    (2011)
  • I.M. Shapiro et al.

    Matrix vesicles: are they anchored exosomes?

    Bone

    (2015)
  • M. Kuzynski et al.

    Dual role of the Trps1 transcription factor in dentin mineralization

    J. Biol. Chem.

    (2014)
  • F. Priam et al.

    New cellular models for tracking the odontoblast phenotype

    Arch. Oral Biol.

    (2005)
  • R. Garimella et al.

    Primary culture of rat growth plate chondrocytes: an in vitro model of growth plate histotype, matrix vesicle biogenesis and mineralization

    Bone

    (2004)
  • H. Cai et al.

    Coats, tethers, Rabs, and SNAREs work together to mediate the intracellular destination of a transport vesicle

    Dev. Cell

    (2007)
  • J.E. Hale et al.

    The mechanism of matrix vesicle formation. Studies on the composition of chondrocyte microvilli and on the effects of microfilament-perturbing agents on cellular vesiculation

    J. Biol. Chem.

    (1987)
  • M. Simons et al.

    Exosomes—vesicular carriers for intercellular communication

    Curr. Opin. Cell Biol.

    (2009)
  • T. Kirsch et al.

    The roles of annexins and types II and X collagen in matrix vesicle-mediated mineralization of growth plate cartilage

    J. Biol. Chem.

    (2000)
  • G. Nangami et al.

    Fetuin-A associates with histones intracellularly and shuttles them to exosomes to promote focal adhesion assembly resulting in rapid adhesion and spreading in breast carcinoma cells

    Exp. Cell Res.

    (2014)
  • K. Tompkins et al.

    Characterization of a mouse amelogenin [A-4]/M59 cell surface receptor

    Bone

    (2006)
  • H. Zhang et al.

    Full length amelogenin binds to cell surface LAMP-1 on tooth root/periodontium associated cells

    Arch. Oral Biol.

    (2010)
  • M. Yanez-Mo et al.

    Biological properties of extracellular vesicles and their physiological functions

    J. Extracell. Vesicles

    (2015)
  • M. Colombo et al.

    Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles

    Annu. Rev. Cell Dev. Biol.

    (2014)
  • C.M. Giachelli et al.

    Regulation of vascular calcification: roles of phosphate and osteopontin

    Circ. Res.

    (2005)
  • K. Johnson et al.

    Linked deficiencies in extracellular PP(i) and osteopontin mediate pathologic calcification associated with defective PC-1 and ANK expression

    J. Bone Miner. Res.

    (2003)
  • G. Luo et al.

    Spontaneous calcification of arteries and cartilage in mice lacking matrix GLA protein

    Nature

    (1997)
  • D.A. Prosdocimo et al.

    Regulation of vascular smooth muscle cell calcification by extracellular pyrophosphate homeostasis: synergistic modulation by cyclic AMP and hyperphosphatemia

    Am. J. Physiol. Cell Physiol.

    (2010)
  • T. Sallam et al.

    Regulatory circuits controlling vascular cell calcification

    Cell. Mol. Life Sci.

    (2013)
  • L.J. Schurgers et al.

    Matrix Gla-protein: the calcification inhibitor in need of vitamin K

    Thromb. Haemost.

    (2008)
  • Z. Szeberin et al.

    Serum fetuin-A levels inversely correlate with the severity of arterial calcification in patients with chronic lower extremity atherosclerosis without renal disease

    Int. Angiol.

    (2011)
  • J. Sodek et al.

    Osteopontin

    Crit. Rev. Oral Biol. Med.

    (2000)
  • D.A. McKnight et al.

    Molecular evolution of dentin phosphoprotein among toothed and toothless animals

    BMC Evol. Biol.

    (2009)
  • B.L. Foster et al.

    Regulation of cementoblast gene expression by inorganic phosphate in vitro

    Calcif. Tissue Int.

    (2006)
  • S. Khoshniat et al.

    The emergence of phosphate as a specific signaling molecule in bone and other cell types in mammals

    Cell. Mol. Life Sci.

    (2011)
  • R. Kumar

    Phosphate sensing

    Curr. Opin. Nephrol. Hypertens.

    (2009)
  • M. Julien et al.

    Phosphate stimulates matrix Gla protein expression in chondrocytes through the extracellular signal regulated kinase signaling pathway

    Endocrinology

    (2007)
  • M. Julien et al.

    Phosphate-dependent regulation of MGP in osteoblasts: role of ERK1/2 and Fra-1

    J. Bone Miner. Res.

    (2009)
  • M.S. Razzaque

    Phosphate and vitamin D in chronic kidney disease

    Contrib. Nephrol.

    (2013)
  • Cited by (47)

    • MV-mediated biomineralization mechanisms and treatments of biomineralized diseases

      2023, Medicine in Novel Technology and Devices
      Citation Excerpt :

      The yield is the ratio of ALP activity to total protein concentration. After extraction, MVs are suspended in PBS and stored at −80 ​°C or under liquid nitrogen [43,49]. The experimental methods of these three sources have their own merits.

    • Calcium and phosphate and their role in matrix vesicles: A biological view

      2023, Mineralizing Vesicles: From Biochemical and Biophysical Properties to Their Roles in Physiology and Disease
    • Physiological biomineralization. The properties and role of matrix vesicles in skeletal and dental calcifications

      2023, Mineralizing Vesicles: From Biochemical and Biophysical Properties to Their Roles in Physiology and Disease
    View all citing articles on Scopus

    These authors equally contributed to this work.

    View full text