Sie sind auf Seite 1von 12

Downloaded from http://perspectivesinmedicine.cshlp.

org/ at NYU MED CTR LIBRARY on April 4, 2018 - Published by Cold Spring
Harbor Laboratory Press

Mechanism of Bone Mineralization

Monzur Murshed1,2,3
1
Faculty of Dentistry, McGill University, Montreal, Quebec H3A 1G1, Canada
2
Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec
H4A 3J1, Canada
3
Shriners Hospital for Children, Montreal, Quebec H4A 0A9, Canada
Correspondence: monzur.murshed@mcgill.ca

Mineralized “hard” tissues of the skeleton possess unique biomechanical properties to


support the body weight and movement and act as a source of essential minerals required
for critical body functions. For a long time, extracellular matrix (ECM) mineralization in the
vertebrate skeleton was considered as a passive process. However, the explosion of genetic
studies during the past decades has established that this process is essentially controlled by
multiple genetic pathways. These pathways regulate the homeostasis of ionic calcium and
inorganic phosphate—two mineral components required for bone mineral formation, the
synthesis of mineral scaffolding ECM, and the maintainence of the levels of the inhibitory
organic and inorganic molecules controlling the process of mineral crystal formation and its
growth. More recently, intracellular enzyme regulators of skeletal tissue mineralization have
www.perspectivesinmedicine.org

been identified. The current review will discuss the key determinants of ECM mineralization
in bone and propose a unified model explaining this process.

T he mineralized extracellular matrix (ECM) is


a unique feature of the vertebrate skeletoden-
tal system. The massive load-bearing capacity of
The origin of biomineralization has been
traced back to the late Precambrian period after
tectonic activities caused a marked increase of
the mineralized skeleton permitted the evolu- soluble minerals in the seawater (Wagner and
tionary emergence of large vertebrates, such as Aspenberg 2011). It is commonly believed that
blue whales, which can weigh up to 180 tons. marine organisms first developed primitive exo-
However, the functions of the mineralized tis- skeletons made up of calcium carbonate and/or
sues are not limited to support the body mass, calcium phosphate minerals (Knoll 2003; Wag-
protect the internal soft organs, or to facilitate ner and Aspenberg 2011). As part of the process
locomotion and mastication only; they also of evolutionary adaptations, the skeletal tissues
serve as a readily accessible reservoir for essen- were internalized, which paved the way for the
tial minerals that are indispensable for many emergence of organisms with larger body sizes.
physiologic activities. The current review will Whereas it is not clear what prompted some
focus on the mechanism of calcium phosphate primitive organisms to deposit calcium phos-
biomineralization in the vertebrate skeleton. phate instead of calcium carbonate minerals in

Editors: Gerard Karsenty and David T. Scadden


Additional Perspectives on Bone: A Regulator of Physiology available at www.perspectivesinmedicine.org
Copyright © 2018 Cold Spring Harbor Laboratory Press; all rights reserved
Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a031229

1
Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on April 4, 2018 - Published by Cold Spring
Harbor Laboratory Press

M. Murshed

their skeletal tissues, it is conceivable that the X-ray diffraction analysis. This pattern suggest-
deposited calcium phosphate minerals provided ed that the precipitated mineral is amorphous
certain physiological advantages. Indeed it has calcium phosphate (ACP), not apatite. Interest-
been suggested that the presence of calcium ingly, when X-ray diffraction analyses were per-
phosphate minerals makes the skeleton more formed on the same samples 2 days later, the
stable under acidic conditions (Wagner and As- precipitates were found to be a poorly formed
penberg 2011). Vertebrate organisms rely on crystalline apatite (Boskey 1997). This initial
ATP generation via anaerobic glycolysis for sud- finding of mineral phase transition in vitro
den rapid movements during “fight-or-flight” prompted follow-up experiments using miner-
situations. The activation of this pathway results alized tissue samples and, subsequently, the
in an acidic tissue environment, which would presence of ACP in the embryonic chick bones
have destabilized a skeleton made up of calcium was reported by the same group (Eanes et al.
carbonate more readily. 1965). However, the controversy on whether
bone contains ACP continued as later studies
concluded that the embryonic bones do not con-
MINERAL COMPOSITION OF BONE
tain ACP (Bonar et al. 1983; Grynpas et al.
In 1771, Scheele first reported the presence of 1984). Since then, more sophisticated analyses
calcium phosphate minerals in bone (Scheele of bone minerals using Raman spectra, Fourier-
1931). Later, bone mineral was described as a transform infrared spectroscopy (FTIR), and
form of hydroxyapatite [(Ca)10 (PO4)6(OH)2] synchrotron-generated X-ray diffraction tech-
comparable to geological apatite (a group of niques have been performed; however, a consen-
phosphate minerals) by both chemical compo- sus on the issue is still elusive (Rey et al. 2009).
sition and X-ray diffraction pattern analyses (see The presence of amorphous mineral phase
www.perspectivesinmedicine.org

Eliaz and Metoki 2017 for a comprehensive re- has been reported in some invertebrates (Becker
view on the historical perspective). However, et al. 1976, 2005). Additionally, ACP has been
subsequent studies demonstrated that minerals detected at the sites of ectopic calcification in the
in bone do not have a uniform composition (Rey vertebrates (Marulanda et al. 2017). In two stud-
et al. 1995; Pasteris et al. 2004). Although there ies published by Mahamid et al. (2008, 2010),
are still some disagreements about the initial ACP has been identified as a major mineral
phase of the deposited mineral and its time-de- phase in zebrafish fin bones, suggesting that
pendent transition to apatite, it is now accepted amorphous to crystalline phase transition oc-
that the matured bone mineral is a substituted curs in vivo. On the other hand, concerns have
crystalline phase of calcium phosphate, referred been raised about this latter finding on the
to as carbonated hydroxyapatite (Mahamid et al. ground of technical limitations, including pos-
2008). High-resolution transmission electron sible mineral phase transition during sample
microscopy (TEM), 3D stereoscopic TEM, and collection and processing (Rey et al. 2009).
atomic force microscopy on organic matrix-free
bone samples provided accurate measurements
DETERMINANTS OF SKELETAL TISSUE
of bone crystals. These studies have firmly estab-
MINERALIZATION
lished that bone crystals are nanosized, long, and
very thin platelets (Rey et al. 2009). In human embryos, although primary ossifica-
The source of the dispute regarding the early tion centers of endochondral bones (e.g., verte-
mineral phase in bone appears to be a simple in brae and long bones) appear between 8 and 12
vitro experiment performed in the mid-sixties in weeks of gestation, the bulk mineralization of
the laboratory of Dr. Posner (Boskey 1997). The skeletal tissues does not occur until the third
mixing of concentrated solutions of calcium trimester (Kovacs 2003). The type I collagen-
chloride and sodium acid phosphate resulted rich ECM in the intramembranous bones min-
in the precipitation of calcium phosphate salts, eralizes directly, whereas in the endochondral
which showed a broad and diffused pattern by bones, mineralization starts concomitantly at

2 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a031229
Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on April 4, 2018 - Published by Cold Spring
Harbor Laboratory Press

Mechanism of Bone Mineralization

two sites: within the core region of the cartilage or XLH patients leads to an increase of circulat-
anlagen and along the bone collar surrounding ing fibroblast growth factor 23 (FGF23) pro-
it (Karsenty and Wagner 2002). In the cartilage, duced by osteoblasts and osteocytes (Xia et al.
the type X collagen matrix synthesized by the 2007). Increased FGF23, in turn, down-regulates
hypertrophic chondrocytes serves as the mineral the expression of sodium phosphate transport-
scaffold, whereas in the bone collar (and later ers in the kidney tubules preventing Pi reabsorp-
in the trabecular bones), the primary mineral tion and causing hypophosphatemia. As expect-
scaffolding protein is the osteoblast-derived ed, in ApoE-Fgf23 transgenic mice in which
type I collagen (Karsenty and Wagner 2002). systemic FGF23 level increases in the absence
A brief discussion on the key determinants reg- of any PHEX mutations, a reduction of systemic
ulating skeletal ECM mineralization is pre- Pi level and osteomalacia comparable to that of
sented below. Hyp mice have been reported (Bai et al. 2004).
The similarity of the osteomalacia traits in ApoE-
Fgf23 and Hyp mice suggests that the accompa-
Systemic Levels of Calcium
nied hypophosphatemia is the main cause of the
and Phosphate Ions
phenotype in these two models.
The chemical structure of bone mineral implies More recently, osteopontin, a protein be-
that the extracellular levels of ionic calcium longing to the family of small integrin-binding
(Ca2+) and inorganic phosphate (Pi) will be ligand, N-linked glycoproteins (SIBLINGs), has
two critical determinants for bone mineraliza- been shown as a substrate for PHEX (Barros
tion. Indeed, data from patients and animal et al. 2013). It has been proposed that in the
models of human diseases clearly demonstrates absence of PHEX, accumulation of osteopontin
that the reduction of systemic Pi levels with or in the ECM may contribute to the “hard” tissue
www.perspectivesinmedicine.org

without any alteration of the Ca2+ levels lead to mineralization defects seen in the XLH patients
osteomalacia with the characteristic increase of and in Hyp mice. Another SIBLING protein,
unmineralized osteoid volume. The importance dentin matrix protein 1 (DMP1), has been
of systemic levels of these mineral ions in bone shown to be an important regulator of circulat-
mineralization was demonstrated by impair- ing FGF23 levels (Martin et al. 2011). As is the
ment of the 1,25 dihydroxy vitamin D3 signaling case with the inactivating mutations in PHEX,
pathway. For example, mutations in 25, hydroxy DMP1 deficiency leads to severe osteomalacia.
D3-1 α-hydroxylase (required for functional vi- A severe osteomalacia phenotype is also seen
tamin D synthesis) or inactivating mutations in in the calcium sensing receptor (CaSR) knock-
vitamin D receptor, impair the absorption of out mice. The ablation of the Casr gene leads to a
Ca2+ and Pi in the gut (Dardenne et al. 2001; marked increase of systemic parathyroid hor-
Masuyama et al. 2001; Panda et al. 2001). Addi- mone (PTH) levels leading to hypercalcemia ac-
tionally, these mutations also reduce the mobi- companied by hypophosphatemia as a result of
lization of these ions from the bone by restricting increased urinary excretion of Pi (Tu et al. 2003).
bone resorption (Suda et al. 1992). Interestingly, the observed increase of Ca2+ in
Although Ca2+ and Pi are both integral parts the circulation failed to prevent osteomalacia
of bone mineral, genetic experiments in mice in these mice. On the other hand, an opposite
suggest that circulating Pi may have a more phenotype has been seen in mice lacking glial
prominent role in the regulation of bone miner- cell missing 2 (GCM2), a transcription factor
alization. For example, in Hyp mice with a mu- required for the development of the parathyroid
tation in the Phex gene (a model of human X- gland (Gunther et al. 2000). The ablation of
linked hypophosphatemia [XLH]), the calcium Gcm2 impairs the development of the parathy-
level is normal, but the systemic Pi level is re- roid gland causing a marked reduction of PTH
duced to almost half, causing a severe osteoma- level in the circulation. A basal PTH level is
lacia phenotype (Eicher et al. 1976; Costa et al. maintained as the thymus acts as a secondary
1981). The inactivation of PHEX in the Hyp mice source for the hormone. As expected, the sur-

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a031229 3
Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on April 4, 2018 - Published by Cold Spring
Harbor Laboratory Press

M. Murshed

viving Gcm2 −/− mice show a reduction of serum copy of the mineralizing bones from newborn
Ca2+ levels and an increase of serum Pi levels. mice by Sheldon and Robinson (1957). Al-
However, despite the significant reduction of though earlier studies provided circumstantial
systemic Ca2+ levels, no osteomalacia was ob- evidence that mineral crystals are deposited
served in these mice (Gunther et al. 2000). within the collagen fibrils, this study first dem-
Currently, a mouse model showing isolated onstrated that there are two sites on the collagen
hypocalcemia with a normal serum Pi level is matrix in bone where mineral deposition starts
unavailable. Considering that Ca2+ is an integral (1) at the intrafibrillar gap spaces, where the car-
component of bone mineral, it is possible that boxy- and amino-terminal ends of two serially
such a model of isolated hypocalcemia, if exist- arranged collagen triple helices meet, and (2)
ed, would have shown impaired bone minerali- interfibrillar spaces between the fibrils (Sheldon
zation. However, phenotypic comparisons of and Robinson 1957). Since then, numerous
the mouse models with abnormal Ca2+ and/or studies have confirmed the critical role of colla-
Pi homeostasis discussed above suggest that the gen matrix as a scaffold for bone mineral
osteomalacia phenotype in a model of isolated deposition. These include the initial studies
hypocalcemia may not be as severe as seen in demonstrating that collagen sponges can be
isolated hypophosphatemia. Because of the crit- mineralized in vitro, implanted collagen can
ical role of Ca2+ in many physiologic activities, mineralize in vivo, and demineralized bone col-
its serum level is more tightly regulated than that lagen can be mineralized under a cell-free con-
of Pi. Moreover, ubiquitously present inorganic dition (Mergenhagen et al. 1960; Bachra and
pyrophosphate (PPi), a potent inhibitor of ECM Fischer 1968a,b). More recently, genetic experi-
mineralization, is a Pi derivative (see below) ments have demonstrated that the reduction of
(Terkeltaub 2001). These observations suggest collagen synthesis in bone results in a reduction
www.perspectivesinmedicine.org

that early organisms depositing calcium phos- of mineralized bone mass (Yang et al. 2004).
phate minerals in the skeletal tissues evolved to The exact mechanism by which the collagen
regulate ECM mineralization by modulating the lattice facilitates mineral deposition is still un-
Pi levels and its incorporation into the growing known. The amino acid side chains exposed at
mineral crystals. the intrafibrillar gap space and at the interfibril-
lar space may regulate this process. However, it
is possible that the dense packaging of collagen
Collagen Scaffold
molecules and their hierarchical organization,
The organic part of the bone ECM is primarily rather than the primary structure of the protein
composed of type I collagen. Two genes, Col1a1 is the driving force underlying the nucleation of
and Col2a1, encode the α1 and α2 chains of type hydroxyapatite. Indeed, one theory attempts to
I collagen, respectively. Two α1 and one α2 explain the mineralization of bone ECM by the
chains assemble together to form the collagen size-exclusion properties of the collagen scaffold
triple helix in the extracellular spaces of the bone (Toroian et al. 2007; Price et al. 2009). Accord-
microenvironment. In a hierarchical fashion, ing to this theory, the nanoscale gaps present
these helices are first arranged axially in a stag- within a collagen fibril and in between the fibrils
gered manner as collagen fibrils, which are then arranged in a fiber allow the access of Ca2+ and
bundled together to form the collagen fibers Pi ions, but not the large proteins, which can
(Shoulders and Raines 2009). In a healthy indi- inhibit the formation and growth of the nascent
vidual, the mineralization of the unmineralized hydroxyapatite crystals inside the scaffold.
collagen (osteoid) occurs seamlessly in contin-
uation of the existing mineralized matrix on
Mineralization Inhibitors
which the newly synthesized osteoid is depos-
ited by the osteoblasts. Considering that the concentrations of various
The involvement of collagen in bone miner- ions in all the tissues of the body are at equilib-
alization came from the initial electron micros- rium to that of blood, it is likely that Ca2+ and Pi

4 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a031229
Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on April 4, 2018 - Published by Cold Spring
Harbor Laboratory Press

Mechanism of Bone Mineralization

levels do not differ significantly between the terase nucleotide pyrophosphatase I (Enpp1)
“soft” and skeletal “hard” tissues. This may raise gene (Koshizuka et al. 2001).
the question—why do some “soft” tissues, such Interestingly, both Ank and Enpp1 genes are
as blood vessels and some cartilaginous tissues, highly expressed in bone, more specifically in the
never mineralize despite their high collagen osteoblasts (Murshed et al. 2005). Although the
content? The most straightforward answer to presence of a mineralization inhibitor in bone
this question can be obtained from several hu- may appear counterintuitive, this quandary can
man diseases of pathologic “soft” tissue miner- be explained by the presence of a strong alkaline
alization and their animal models. For example, phosphatase (ALPL) activity in bone. The car-
generalized arterial calcification in infancy dinal role of ALPL during skeletal mineraliza-
(GACI) and progressive ankylosis in humans tion in humans was demonstrated by many dif-
are both caused by the reduced level of PPi in ferent mutations identified in the ALPL gene in
the bone joints (Ho et al. 2000; Albright et al. hypophosphatasia patients, causing a wide range
2015). PPi is a mineralization inhibitor that is of phenotypic severity. Some of these patients
known for its potent antimineralization proper- show very severe osteomalacia and fetal/perina-
ties for over 50 years (Fleisch and Bisaz 1962). tal lethality, while others show milder, progres-
A wide distribution of the PPi synthesis and/ sive osteomalacia later in life (Mornet 2000; Tail-
or extracellular transport machinery in verte- landier et al. 2000, 2001). The genetic models
brate organisms prevents pathologic “soft” tis- lacking ALPL activity further confirmed the re-
sue mineralization. quirement of this enzyme for bone mineraliza-
PPi is composed of two inorganic Pi groups tion (Fedde et al. 1999; Anderson et al. 2004).
joined by an ester linkage (Terkeltaub 2001). ALPL-mediated hydrolysis of PPi has two
This inorganic mineralization inhibitor is pro- implications—first, it reduces the amount of
www.perspectivesinmedicine.org

duced both intracellularly and extracellularly PPi in the bone microenvironment and, second,
via enzymatic reactions. Intracellularly, PPi it increases the amount of mineralization-pro-
can be generated as a byproduct of enzymatic moting Pi ions by liberating them from PPi.
activities in numerous metabolic pathways. In- These coupled activities alter the Pi/PPi ratio in
tracellular PPi is transported to the extracellular the bone ECM in such a way that the formation
space through a transmembrane transporter and growth of the apatite crystal is promoted
called ANK (Ho et al. 2000). On the other (Hessle et al. 2002; Murshed et al. 2005). The
hand, extracellularly, a direct cleavage of the critical effect of this ratio in regulating ECM
phosphodiester bond in purine and pyrimidine mineralization has been demonstrated in ank/
nucleoside triphosphates (e.g., ATP) by nucle- ank;Hyp compound-mutant mice, which show
otide pyrophosphatase/phosphodiesterase en- a marked reduction of joint mineralization
zymes can also generate PPi (Hessle et al. (Murshed et al. 2005). The low extracellular Pi/
2002). PPi ratio in these mice in comparison to the
At a threshold concentration, PPi prevents original ank/ank mice prevented the ectopic
the incorporation of Ca2+ and Pi into nascent mineral deposition.
apatitic crystals and inhibits their growth (Ter- The ALPL “knockout” mice were used to
keltaub 2001). Studies on the genetic models validate the therapeutic effectiveness of a genet-
have identified the key proteins involved in ically engineered ALPL molecule with the ability
PPi-mediated inhibition of pathologic calcifica- to bind to the bone matrix in vivo. These mice
tion. In a mouse model of human progressive showed a remarkable improvement of the bone
ankylosis with homozygous mutations in the phenotype, as the hypophosphatasia-associated
Ank gene (ank/ank), massive mineral deposi- osteomalacia was prevented (Yadav et al. 2011a).
tion is seen in the joints and other soft tissues This approach of enzyme-replacement therapy
(Ho et al. 2000). A similar phenotype has been has been successfully used to treat the hypo-
seen in ttw/ttw mice, which carry homozygous phosphatasia-associated skeletal abnormalities
mutations in the ectonucleotide phosphodies- in human clinical trials (Kitaoka et al. 2017).

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a031229 5
Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on April 4, 2018 - Published by Cold Spring
Harbor Laboratory Press

M. Murshed

Matrix Gla protein (MGP) and fetuin, two head revealed that the phenotype is caused by
proteins involved in the prevention of “soft” tis- the impaired growth of the maxillary and pala-
sue mineralization are also present in the skeletal tine bones in comparison to the mandibular
hard tissues (Jahnen-Dechent et al. 1997). How- bones. Marulanda et al. (2017) reported that
ever, considering that fetuin “knockout” mice do the hypoplastic midface was not a result of cra-
not show any overt bone mineralization defects niosynostosis as seen in several other mouse
or skeletal anomalies, the current discussion will models of midface hypoplasia, but a result of
focus on MGP only. In humans, the homozygous ectopic mineralization of the cartilaginous part
loss-of-function mutations in MGP leads to a of the nasal septum, which normally does not
rare genetic disorder known as Keutel syndrome mineralize. This abnormal mineralization in-
(Fryns et al. 1984; Munroe et al. 1999). The pa- duced apoptosis of the chondrocytes present
tients show abnormal mineralization of their in the nasal septum (Marulanda et al. 2017).
cartilaginous tissues leading to midface hypopla- The loss of the matrix synthesizing active chon-
sia, shortening of terminal phalanges, tracheo- drocytes during the early growth phase and the
bronchial calcification, and vascular abnormali- increased rigidity of the ECM upon mineraliza-
ties, including calcification of the arterial media. tion might have impaired the growth of the nasal
MGP-deficient mice recapitulated all of these septum and that of the maxillary complex. In-
anomalies albeit with a more severe vascular cal- terestingly, the deposited minerals in the septal
cification phenotype (Luo et al. 1997). cartilage of the mutant mice show the presence
MGP, a small mineral-binding protein is of ACP, suggesting that MGP-mediated inhibi-
highly expressed by chondrocytes and vascular tion of mineralization occurs during the early
smooth muscle cells (Luo et al. 1995, 1997). Al- phases of mineral precipitation.
though MGP was initially purified from the bo-
www.perspectivesinmedicine.org

vine bones, its expression is markedly lower in


THE ROLE OF INTRACELLULAR ENZYMES IN
the trabecular bones in comparison to the pre-
SKELETAL MINERALIZATION
hypertrophic zone of the developing growth
plate (Price and Williamson 1985; Luo et al. Whereas most of the published work on the
1995). This expression pattern suggests that mechanism of cartilage and bone mineralization
MGP present in bone might have originated focuses primarily on the extracellular determi-
elsewhere (e.g., in the vascular tissues) and nants, newer studies have identified several in-
transported to bone via blood. Transgenic over- tracellular enzymes as important regulators of
expression of Mgp in the osteoblasts resulted in a this process. The main strength of these studies
moderate level of osteomalacia affecting both is the use of in vivo gene ablation models exhib-
intramembranous and endochondral bones iting obvious “hard” tissue mineralization de-
(Murshed et al. 2004). This observation high- fects, which are not caused by the alterations
lights the importance of the mechanisms to con- of the known determinants discussed above.
trol the expression of mineralization inhibitors The intracellular enzymes, sphingomyelin phos-
in the mineralizing skeletal tissues. phodiesterase 3 (SMPD3) and phosphatase or-
In agreement with the observation that Mgp phan 1 (PHOSPHO1) reported by these studies
is weakly expressed in bone in comparison to the are both involved in the metabolism of phos-
cartilaginous tissues, the whole-body ablation of pholipids and/or associated metabolites (Aubin
Mgp in mice does not overtly affect bone min- et al. 2005; Macrae et al. 2010).
eralization, but shows skeletal anomalies associ- SMPD3 (also known as neutral sphingo-
ated with abnormal cartilage mineralization myelinase 2) is a cell membrane–bound lipid-
(Marulanda et al. 2017). The most prominent metabolizing enzyme expressed in high amounts
of these anomalies is midface hypoplasia, which in the brain, cartilage, and bone. Among various
is also seen in the patients with Keutel syn- acid, basic, and neutral sphingomyelinases, only
drome. Cephalometric analyses of the micro- SMPD3 deficiency leads to impaired minerali-
computed tomography (CT) images of the zation of the skeletal tissues (Stoffel et al. 2005,

6 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a031229
Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on April 4, 2018 - Published by Cold Spring
Harbor Laboratory Press

Mechanism of Bone Mineralization

2007; Macrae et al. 2010; Khavandgar and mineralization defects (Holland et al. 2007).
Murshed 2015). SMPD3 cleaves sphingomyelin These findings lead to two conclusions: it is pos-
present in the cell membrane to generate cer- sible that the ceramides generated from these
amide, a bioactive lipid molecule, and phospho- two pathways contribute to different pools
choline, an essential nutrient. Emerging data with distinct cellular functions; alternatively, it
from the analysis of genetically altered mouse is possible that ceramide does not play a role in
models suggest that SMPD3 activity during em- the process of bone mineralization. While at
bryonic development plays a critical role in present, no experimental data directly suggests
normal skeletogenesis. This insight originally the involvement of ceramide in ECM minerali-
came from two different mouse models lacking zation, the importance of phosphocholine in
SMPD3. One of these models, fro/fro, carries a this process has been convincingly shown by
chemically induced deletion encompassing animal experiments (Macrae et al. 2010; Yadav
part of intron 8 and most of exon 9 of the et al. 2011b). Phosphocholine generated from
Smpd3 gene, whereas the other mouse model sphingomyelin by SMPD3, or from dietary cho-
(Smpd3 −/−) was generated by the conventional line by two isoforms of choline kinases, can be
gene-targeting approach (Aubin et al. 2005; cleaved by PHOSPHO1, an intracellular enzyme
Stoffel et al. 2005, 2007). with phosphatase activity. The deficiency of
The fro mutation completely abolishes the PHOSPHO1 in mice has been shown to cause
enzymatic activity of SMPD3, but does not affect similar bone mineralization defects as seen in
its membrane localization (Khavandgar et al. fro/fro mice (Yadav et al. 2011b).
2011). A severe mineralization defect affecting
both intramembranous and endochondral
INTRACELLULAR REGULATORS OF ECM
bones, and an abnormal delay of apoptosis of
www.perspectivesinmedicine.org

MINERALIZATION AND THE “MATRIX


the hypertrophic chondrocytes during the early
VESICLE THEORY”
stages of skeletal development are the hallmarks
of the skeletal phenotypes in fro/fro mice. Re- Whereas the regulatory roles of two intracellular
cently, Li et al. have demonstrated that SMPD3 enzymes, SMPD3 and PHOSPHO1, in ECM
activity in both chondrocytes and osteoblasts are mineralization are now well established, the
required for a normal bone development (Li question remains how these intracellular en-
et al. 2016). The poor mineralization of the skel- zymes may regulate a process that occurs outside
etal tissues in fro/fro mice is seen without any the cells. A possible mechanism may involve
alterations of the homeostasis of Ca2+, Pi, and matrix vesicles (MVs), which are nanosized
PPi (Khavandgar et al. 2011). This observation (20–200 nm) vesicles released by the cells in a
suggests that the loss of SMPD3 function affects mineralizing tissue (Anderson et al. 2004, 2005;
ECM mineralization through a novel, yet un- Golub 2009; Wu et al. 2002). Since their initial
known, mechanism. discoveries as chondrocyte-derived vesicular
As mentioned above, SMPD3 cleaves sphin- bodies promoting the initiation of mineral nu-
gomyelin to generate many different species of cleation, there have been considerable efforts to
ceramides that regulate a myriad of cell func- explain various types of ECM mineralization,
tions. SMPD3 activity also liberates an essential including pathologic mineralization of “soft tis-
nutrient, phosphocholine. Whereas the role of sues” using the “matrix vesicle theory” (Ander-
ceramide is well documented in the regulation of son 1984). Although there is no general consen-
apoptosis (Obeid et al. 1993), it is not yet clear sus, it has been suggested that MVs provide an
whether both ceramide and phosphocholine or isolated microenvironment to facilitate the ini-
only one of these metabolites is involved in skel- tial nucleation of apatite mineral. Mineral crys-
etal tissue mineralization. Interestingly, gene tals formed inside the MVs grow progressively
knockout experiments reducing ceramide bio- in size by the addition of Ca2+ and Pi ions and
synthesis via an alternative pathway without in- eventually rupture the MV membrane to be de-
volving SMPD3 activity did not report any ECM posited on the collagen scaffold.

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a031229 7
Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on April 4, 2018 - Published by Cold Spring
Harbor Laboratory Press

M. Murshed

Wu et al. (2002) showed extensive phospho- SMPD3 may generate additional phosphocho-
lipid degradation in the mineralizing MVs with line to be cleaved by PHOSPHO1. Although
a concomitant increase of free fatty acids. This further studies will be needed to establish that
observation suggested the presence of phospho- SMPD3 and PHOSPHO1 works in a relay in the
lipase activity inside the MVs and demonstrated MVs, such a possibility is supported by the ob-
a link between phospholipid metabolism and servation that both PHOSPHO1 and SMPD3
the initiation of ECM mineralization. Phospho- are present in the MV preparations (Mebarek
lipase activity generates phosphocholine and et al. 2013). Apart from its role described above,
phosphoethanolamine, which can be cleaved SMPD3 might be involved in the biogenesis of
by PHOSPHO1 releasing free Pi inside the the MVs.
MVs. The increase of Pi inside the MVs may
alter the Pi/PPi ratio to favor the seeding of early
SUMMARY
mineral crystals.
Being a membrane-bound enzyme with an Based on the discussion above, a unified model
intracellular catalytic domain, SMPD3 is expect- for bone mineralization can be proposed. When
ed to be present inside the MVs. The cleavage of present at physiologic concentrations, two min-
sphingomyelin present in the MV membrane by eral ions—Ca2+ and Pi—will promote hydroxy-
www.perspectivesinmedicine.org

3
ALPL

PD
SM

SM
PPi
2Pi
Ca2+
Phosphocholine
PHOSPHO1

Ca2+ Pi + Choline

HA
Collagen

Figure 1. Model of bone mineralization. Serum calcium (Ca2+), inorganic phosphate (Pi) levels, and a mineral
scaffolding collagen-rich extracellular matrix (ECM) are important determinants of bone mineralization. Alka-
line phosphatase (ALPL), an ectoenzyme tethered to the osteoblast cell membrane, cleaves inorganic pyrophos-
phate (PPi), a small, but potent mineralization inhibitor. This facilitates bone ECM mineralization in two ways:
first, it reduces the level of a mineralization inhibitor, and second, in the process generates Pi, an activator of ECM
mineralization. This coupled ALPL activity alters the Pi/PPi ratio in the bone microenvironment to favor bone
mineralization. Compact hierarchical assembly of collagen molecules in the fibrils and fibers results in both intra-
and interfibrillar nanoscale gaps. These gaps are accessible by Ca2+ and Pi ions, but not by the large protein
inhibitors of ECM mineralization. This may explain why there are mineral deposits both inside and in the gaps in
between the collagen fibrils. Matrix vesicle (MV)–mediated mineralization may serve as an auxiliary mechanism
for bone mineralization. These nanoscale vesicles carrying the intracellular mineralization-promoting enzymes
bud off from the mineralizing cells. Enzymes like SMPD3 and phospholipases present inside the MVs may cleave
the phospholipids (e.g., sphingomyelin [SM]) to generate phosphocholine, which in turn can be cleaved by
another cytosolic enzyme PHOSPHO1 liberating free Pi. An increase of intravesicular Pi leads to its precipitation
with Ca2+ to form the nascent hydroxyapatite (HA) crystals.

8 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a031229
Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on April 4, 2018 - Published by Cold Spring
Harbor Laboratory Press

Mechanism of Bone Mineralization

apatite crystal growth within and between the Bachra BN, Fischer HR. 1968a. Mineral deposition in colla-
gen in vitro. Calcif Tissue Res 2: 343–352.
newly synthesized collagen fibrils in the skeletal
Bachra BN, Fischer HR. 1968b. Recalcification of decalcified
ECM. PPi, a chemical derivative of Pi, can inhibit bone collagen in vitro as a model for biologic calcification.
the mineralization process. The presence of a Calcif Tissue Res 2: 7.
scaffolding matrix and a defined extracellular Bai X, Miao D, Li J, Goltzman D, Karaplis AC. 2004. Trans-
ratio of Pi to PPi are two critical determinants genic mice overexpressing human fibroblast growth factor
23 (R176Q) delineate a putative role for parathyroid hor-
of ECM mineralization. ALPL, an ectoenzyme mone in renal phosphate wasting disorders. Endocrinol-
bound to the osteoblast cell membrane, cleaves ogy 145: 5269–5279.
PPi to generate Pi and alters the local Pi to PPi Barros NM, Hoac B, Neves RL, Addison WN, Assis DM,
ratio to favor mineral precipitation (Fig. 1). Murshed M, Carmona AK, McKee MD. 2013. Proteolytic
processing of osteopontin by PHEX and accumulation of
The above model suggests that the specificity
osteopontin fragments in Hyp mouse bone, the murine
of skeletal mineralization can be explained, in model of X-linked hypophosphatemia. J Bone Miner Res
part, by the unique coexpression of tissue-non- 28: 688–699.
specific genes encoding type I collagen and Becker GL, Termine JD, Eanes ED. 1976. Comparative stud-
ALPL. Indeed as in vivo proof for this hypoth- ies of intra- and extramitochondrial calcium phosphates
from the hepatopancreas of the blue crab (Callinectes
esis, it was demonstrated that when the PPi sapidus). Calcif Tissue Res 21: 105–113.
cleaving enzyme, ALPL, was mis-expressed in Becker A, Ziegler A, Epple M. 2005. The mineral phase in the
the dermis and arterial media, tissues rich in cuticles of two species of Crustacea consists of magnesium
fibrillar collagen, it caused rapid ECM mineral- calcite, amorphous calcium carbonate, and amorphous
calcium phosphate. Dalton Trans 10: 1814–1820.
ization (Murshed et al. 2005). However, it
Bonar LC, Roufosse AH, Sabine WK, Grynpas MD,
should be noted that this finding does not rule Glimcher MJ. 1983. X-ray diffraction studies of the crys-
out the existence of auxiliary mechanisms work- tallinity of bone mineral in newly synthesized and density
ing together to mineralize the skeletal ECM. The fractionated bone. Calcif Tissue Int 35: 202–209.
www.perspectivesinmedicine.org

possibility of additional mechanism(s) is sup- Boskey AL. 1997. Amorphous calcium phosphate: The con-
tention of bone. J Dent Res 76: 1433–1436.
ported by the fact that both SMPD3- and Costa T, Marie PJ, Scriver CR, Cole DE, Reade TM, Nogrady
PHOSPHO1-deficient mice show bone miner- B, Glorieux FH, Delvin EE. 1981. X-linked hypophospha-
alization defects. These enzymes may work in temia: Effect of calcitriol on renal handling of phosphate,
concert to increase the Pi levels to facilitate the serum phosphate, and bone mineralization. J Clin Endo-
crinol Metab 52: 463–472.
nucleation of minerals inside the MVs (Fig. 1).
Dardenne O, Prud’homme J, Arabian A, Glorieux FH, St-
Arnaud R. 2001. Targeted inactivation of the 25-hydrox-
yvitamin D(3)-1(α)-hydroxylase gene (CYP27B1) creates
REFERENCES an animal model of pseudovitamin D-deficiency rickets.
Endocrinology 142: 3135–3141.
Albright RA, Stabach P, Cao W, Kavanagh D, Mullen I, Eanes ED, Gillessen IH, Posner AS. 1965. Intermediate states
Braddock AA, Covo MS, Tehan M, Yang G, Cheng Z, et in the precipitation of hydroxyapatite. Nature 208: 365–
al. 2015. ENPP1-Fc prevents mortality and vascular cal- 367.
cifications in rodent model of generalized arterial calcifi-
Eicher EM, Southard JL, Scriver CR, Glorieux FH. 1976.
cation of infancy. Nat Commun 6: 10006.
Hypophosphatemia: Mouse model for human familial
Anderson HC. 1984. Mineralization by matrix vesicles. Scan hypophosphatemic (vitamin D-resistant) rickets. Proc
Electron Microsc 2: 953–964. Natl Acad Sci 73: 4667–4671.
Anderson HC, Sipe JB, Hessle L, Dhanyamraju R, Atti E, Eliaz N, Metoki N. 2017. Calcium phosphate bioceramics: A
Camacho NP, Millán JL, Dhamyamraju R. 2004. Impaired review of their history, structure, properties, coating tech-
calcification around matrix vesicles of growth plate and
nologies and biomedical applications. Materials (Basel)
bone in alkaline phosphatase-deficient mice. Am J Pathol
10: E334.
164: 841–847.
Fedde KN, Blair L, Silverstein J, Coburn SP, Ryan LM, Wein-
Anderson HC, Garimella R, Tague SE. 2005. The role of
matrix vesicles in growth plate development and biomin- stein RS, Waymire K, Narisawa S, Millán JL, MacGregor
eralization. Front Biosci 10: 822–837. GR, et al. 1999. Alkaline phosphatase knock-out mice
recapitulate the metabolic and skeletal defects of infantile
Aubin I, Adams CP, Opsahl S, Septier D, Bishop CE, Auge N, hypophosphatasia. J Bone Miner Res 14: 2015–2026.
Salvayre R, Negre-Salvayre A, Goldberg M, Guénet JL, et
al. 2005. A deletion in the gene encoding sphingomyelin Fleisch H, Bisaz S. 1962. Mechanism of calcification: Inhib-
phosphodiesterase 3 (Smpd3) results in osteogenesis and itory role of pyrophosphate. Nature 195: 911.
dentinogenesis imperfecta in the mouse. Nat Genet 37: Fryns JP, van Fleteren A, Mattelaer P, van den Berghe H.
803–805. 1984. Calcification of cartilages, brachytelephalangy and

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a031229 9
Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on April 4, 2018 - Published by Cold Spring
Harbor Laboratory Press

M. Murshed

peripheral pulmonary stenosis. Confirmation of the Keu- Luo G, D’Souza R, Hogue D, Karsenty G. 1995. The matrix
tel syndrome. Eur J Pediatr 142: 201–203. Gla protein gene is a marker of the chondrogenesis cell
Golub EE. 2009. Role of matrix vesicles in biomineralization. lineage during mouse development. J Bone Miner Res 10:
Biochim Biophys Acta 1790: 1592–1598. 325–334.
Grynpas MD, Bonar LC, Glimcher MJ. 1984. Failure to de- Luo G, Ducy P, McKee MD, Pinero GJ, Loyer E, Behringer
tect an amorphous calcium-phosphate solid phase in RR, Karsenty G. 1997. Spontaneous calcification of arter-
bone mineral: A radial distribution function study. Calcif ies and cartilage in mice lacking matrix GLA protein.
Tissue Int 36: 291–301. Nature 386: 78–81.
Gunther T, Chen ZF, Kim J, Priemel M, Rueger JM, Amling Macrae VE, Davey MG, McTeir L, Narisawa S, Yadav MC,
M, Moseley JM, Martin TJ, Anderson DJ, Karsenty G. Millan JL, Farquharson C. 2010. Inhibition of PHOS-
2000. Genetic ablation of parathyroid glands reveals an- PHO1 activity results in impaired skeletal mineralization
other source of parathyroid hormone. Nature 406: 199– during limb development of the chick. Bone 46: 1146–
203. 1155.
Hessle L, Johnson KA, Anderson HC, Narisawa S, Sali A, Mahamid J, Sharir A, Addadi L, Weiner S. 2008. Amorphous
Goding JW, Terkeltaub R, Millan JL. 2002. Tissue-non- calcium phosphate is a major component of the forming
specific alkaline phosphatase and plasma cell membrane fin bones of zebrafish: Indications for an amorphous pre-
glycoprotein-1 are central antagonistic regulators of bone cursor phase. Proc Natl Acad Sci 105: 12748–12753.
mineralization. Proc Natl Acad Sci 99: 9445–9449. Mahamid J, Aichmayer B, Shimoni E, Ziblat R, Li C, Siegel S,
Ho AM, Johnson MD, Kingsley DM. 2000. Role of the mouse Paris O, Fratzl P, Weiner S, Addadi L. 2010. Mapping
ank gene in control of tissue calcification and arthritis. amorphous calcium phosphate transformation into crys-
Science 289: 265–270. talline mineral from the cell to the bone in zebrafish fin
Holland WL, Brozinick JT, Wang LP, Hawkins ED, Sargent rays. Proc Natl Acad Sci 107: 6316–6321.
KM, Liu Y, Narra K, Hoehn KL, Knotts TA, Siesky A, et al. Martin A, Liu S, David V, Li H, Karydis A, Feng JQ, Quarles
2007. Inhibition of ceramide synthesis ameliorates gluco- LD. 2011. Bone proteins PHEX and DMP1 regulate fibro-
corticoid-, saturated-fat-, and obesity-induced insulin re- blastic growth factor Fgf23 expression in osteocytes
sistance. Cell Metab 5: 167–179. through a common pathway involving FGF receptor
Jahnen-Dechent W, Schinke T, Trindl A, Müller-Esterl W, (FGFR) signaling. FASEB J 25: 2551–2562.
Sablitzky F, Kaiser S, Blessing M. 1997. Cloning and tar- Marulanda J, Eimar H, McKee MD, Berkvens M, Nelea V,
www.perspectivesinmedicine.org

geted deletion of the mouse fetuin gene. J Biol Chem 272: Roman H, Borrás T, Tamimi F, Ferron M, Murshed M.
31496–31503. 2017. Matrix Gla protein deficiency impairs nasal septum
Karsenty G, Wagner EF. 2002. Reaching a genetic and mo- growth, causing midface hypoplasia. J Biol Chem 292:
lecular understanding of skeletal development. Dev Cell 2: 11400–11412.
389–406. Masuyama R, Nakaya Y, Tanaka S, Tsurukami H, Nakamura
Khavandgar Z, Murshed M. 2015. Sphingolipid metabolism T, Watanabe S, Yoshizawa T, Kato S, Suzuki K. 2001.
and its role in the skeletal tissues. Cell Mol Life Sci 72: Dietary phosphorus restriction reverses the impaired
959–969. bone mineralization in vitamin D receptor knockout
Khavandgar Z, Poirier C, Clarke CJ, Li J, Wang N, McKee mice. Endocrinology 142: 494–497.
MD, Hannun YA, Murshed M. 2011. A cell-autonomous Mebarek S, Abousalham A, Magne D, Do le D, Bandoro-
requirement for neutral sphingomyelinase 2 in bone min- wicz-Pikula J, Pikula S, Buchet R. 2013. Phospholipases of
eralization. J Cell Biol 194: 277–289. mineralization competent cells and matrix vesicles: Roles
Kitaoka T, Tajima T, Nagasaki K, Kikuchi T, Yamamoto K, in physiological and pathological mineralizations. Int J
Michigami T, Okada S, Fujiwara I, Kokaji M, Mochizuki Mol Sci 14: 5036–5129.
H, et al. 2017. Safety and efficacy of treatment with asfo- Mergenhagen SE, Martin GR, Rizzo AA, Wright DN, Scott
tase alfa in patients with hypophosphatasia: Results from DB. 1960. Calcification in vivo of implanted collagen.
a Japanese clinical trial. Clin Endocrinol (Oxf ) 87: 10–19. Biochim Biophys Acta 43: 563–565.
Knoll AH. 2003. Biomineralization and evolutionary history. Mornet E. 2000. Hypophosphatasia: The mutations in the
Rev Mineral Geochem 54: 329–356. tissue-nonspecific alkaline phosphatase gene. Hum Mu-
Koshizuka Y, Ikegawa S, Sano M, Nakamura K, Nakamura Y. tat 15: 309–315.
2001. Isolation of novel mouse genes associated with ec- Munroe PB, Olgunturk RO, Fryns JP, Van Maldergem L,
topic ossification by differential display method using ttw, Ziereisen F, Yuksel B, Gardiner RM, Chung E. 1999. Mu-
a mouse model for ectopic ossification. Cytogenet Cell tations in the gene encoding the human matrix Gla pro-
Genet 94: 163–168. tein cause Keutel syndrome. Nat Genet 21: 142–144.
Kovacs CS. 2003. Skeletal physiology: Fetus and neonate. In Murshed M, Schinke T, McKee MD, Karsenty G. 2004. Ex-
Primer on the metabolic bone diseases and disorders of tracellular matrix mineralization is regulated locally; dif-
mineral metabolism. American Society for Bone and ferent roles of two gla-containing proteins. J Cell Biol 165:
Mineral Research, Washington, DC. 625–630.
Li J, Manickam G, Ray S, Oh CD, Yasuda H, Moffatt P, Murshed M, Harmey D, Millan JL, McKee MD, Karsenty G.
Murshed M. 2016. Smpd3 expression in both chondro- 2005. Unique coexpression in osteoblasts of broadly ex-
cytes and osteoblasts is required for normal endochondral pressed genes accounts for the spatial restriction of ECM
bone development. Mol Cell Biol 36: 2282–2299. mineralization to bone. Genes Dev 19: 1093–1104.

10 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a031229
Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on April 4, 2018 - Published by Cold Spring
Harbor Laboratory Press

Mechanism of Bone Mineralization

Obeid LM, Linardic CM, Karolak LA, Hannun YA. 1993. 298-2A>G, T117N, A159T, R229S, 997+2T>A, E274X,
Programmed cell death induced by ceramide. Science A331T, H364R, D389G, 1256delC, R433H, N461I,
259: 1769–1771. C472S) in the tissue-nonspecific alkaline phosphatase
Panda DK, Miao D, Tremblay ML, Sirois J, Farookhi R, (TNSALP) gene in patients with hypophosphatasia.
Hendy GN, Goltzman D. 2001. Targeted ablation of the Hum Mutat 15: 293.
25-hydroxyvitamin D 1α-hydroxylase enzyme: Evidence Taillandier A, Lia-Baldini AS, Mouchard M, Robin B, Muller
for skeletal, reproductive, and immune dysfunction. Proc F, Simon-Bouy B, Serre JL, Bera-Louville A, Bonduelle M,
Natl Acad Sci 98: 7498–7503. Eckhardt J, et al. 2001. Twelve novel mutations in the
Pasteris JD, Wopenka B, Freeman JJ, Rogers K, Valsami- tissue-nonspecific alkaline phosphatase gene (ALPL) in
Jones E, van der Houwen JA, Silva MJ. 2004. Lack of patients with various forms of hypophosphatasia. Hum
OH in nanocrystalline apatite as a function of degree of Mutat 18: 83–84.
atomic order: Implications for bone and biomaterials. Terkeltaub RA. 2001. Inorganic pyrophosphate generation
Biomaterials 25: 229–238. and disposition in pathophysiology. Am J Physiol Cell
Price PA, Williamson MK. 1985. Primary structure of bovine Physiol 281: C1–C11.
matrix Gla protein, a new vitamin K-dependent bone Toroian D, Lim JE, Price PA. 2007. The size exclusion char-
protein. J Biol Chem 260: 14971–14975. acteristics of type I collagen: Implications for the role of
Price PA, Toroian D, Lim JE. 2009. Mineralization by inhib- noncollagenous bone constituents in mineralization. J
itor exclusion: The calcification of collagen with fetuin. J Biol Chem 282: 22437–22447.
Biol Chem 284: 17092–17101. Tu Q, Pi M, Karsenty G, Simpson L, Liu S, Quarles LD. 2003.
Rey C, Miquel JL, Facchini L, Legrand AP, Glimcher MJ. Rescue of the skeletal phenotype in CasR-deficient mice
1995. Hydroxyl groups in bone mineral. Bone 16: 583– by transfer onto the Gcm2 null background. J Clin Invest
586. 111: 1029–1037.
Rey C, Combes C, Drouet C, Glimcher MJ. 2009. Bone min- Wagner DO, Aspenberg P. 2011. Where did bone come
eral: Update on chemical composition and structure. Os- from? Acta Orthop 82: 393–398.
teoporos Int 20: 1013–1021. Wu LN, Genge BR, Kang MW, Arsenault AL, Wuthier RE.
Scheele CW. 1931. The collected papers of Carl Wilhelm 2002. Changes in phospholipid extractability and compo-
Scheele (translated by Leonard Dobbin). G. Bell & Sons, sition accompany mineralization of chicken growth plate
London. cartilage matrix vesicles. J Biol Chem 277: 5126–5133.
www.perspectivesinmedicine.org

Sheldon H, Robinson RA. 1957. Electron microscope studies Xia W, Meng X, Jiang Y, Li M, Xing X, Pang L, Wang O, Pei
of crystal-collagen relationships in bone. IV: The occur- Y, Yu LY, Sun Y, et al. 2007. Three novel mutations of the
rence of crystals within collagen fibrils. J Biophys Biochem PHEX gene in three Chinese families with X-linked dom-
Cytol 3: 1011–1016. inant hypophosphatemic rickets. Calcif Tissue Int 81:
Shoulders MD, Raines RT. 2009. Collagen structure and 415–420.
stability. Annu Rev Biochem 78: 929–958. Yadav MC, Lemire I, Leonard P, Boileau G, Blond L, Beliveau
Stoffel W, Jenke B, Block B, Zumbansen M, Koebke J. 2005. M, Cory E, Sah RL, Whyte MP, Crine P, et al. 2011a. Dose
Neutral sphingomyelinase 2 (smpd3) in the control of response of bone-targeted enzyme replacement for mu-
postnatal growth and development. Proc Natl Acad Sci rine hypophosphatasia. Bone 49: 250–256.
102: 4554–4559. Yadav MC, Simão AM, Narisawa S, Huesa C, McKee MD,
Stoffel W, Jenke B, Holz B, Binczek E, Günter RH, Knifka J, Farquharson C, Millán JL. 2011b. Loss of skeletal miner-
Koebke J, Niehoff A. 2007. Neutral sphingomyelinase alization by the simultaneous ablation of PHOSPHO1
(SMPD3) deficiency causes a novel form of chondrodys- and alkaline phosphatase function: A unified model of
plasia and dwarfism that is rescued by Col2A1-driven the mechanisms of initiation of skeletal calcification. J
smpd3 transgene expression. Am J Pathol 171: 153–161. Bone Miner Res 26: 286–297.
Suda T, Takahashi N, Abe E. 1992. Role of vitamin D in bone Yang X, Matsuda K, Bialek P, Jacquot S, Masuoka HC,
resorption. J Cell Biochem 49: 53–58. Schinke T, Li L, Brancorsini S, Sassone-Corsi P, Townes
Taillandier A, Cozien E, Muller F, Merrien Y, Bonnin E, TM, et al. 2004. ATF4 is a substrate of RSK2 and an
Fribourg C, Simon-Bouy B, Serre JL, Bieth E, Brenner essential regulator of osteoblast biology; implication for
R, et al. 2000. Fifteen new mutations (-195C>T, L-12X, Coffin–Lowry syndrome. Cell 117: 387–398.

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Med doi: 10.1101/cshperspect.a031229 11
Downloaded from http://perspectivesinmedicine.cshlp.org/ at NYU MED CTR LIBRARY on April 4, 2018 - Published by Cold Spring
Harbor Laboratory Press

Mechanism of Bone Mineralization


Monzur Murshed

Cold Spring Harb Perspect Med published online April 2, 2018

Subject Collection Bone: A Regulator of Physiology

Osteoimmunology Mechanism of Bone Mineralization


Kazuo Okamoto and Hiroshi Takayanagi Monzur Murshed
Bone Remodeling and the Microbiome Neural Regulation of Bone and Bone Marrow
Roberto Pacifici Maria Maryanovich, Shoichiro Takeishi and Paul S.
Frenette
Regulation of Bone Remodeling by Parathyroid Regulation of Bone Metabolism by Sex Steroids
Hormone Sundeep Khosla and David G. Monroe
Marc N. Wein and Henry M. Kronenberg
Multiple Myeloma and Bone: The Fatal Interaction The Biology of Bone Metastasis
Silvia Marino and G. David Roodman Mark Esposito, Theresa Guise and Yibin Kang
The Bone Marrow Microenvironment in Health and Biology of Bone: The Vasculature of the Skeletal
Myeloid Malignancy System
Marta Galán-Díez, Álvaro Cuesta-Domínguez and Emma C. Watson and Ralf H. Adams
Stavroula Kousteni
Regulation of Energy Metabolism by Biology of Fibroblast Growth Factor 23: From
Bone-Derived Hormones Physiology to Pathology
Paula Mera, Mathieu Ferron and Ioanna Mosialou Marie Courbebaisse and Beate Lanske

For additional articles in this collection, see http://perspectivesinmedicine.cshlp.org/cgi/collection/

Copyright © 2018 Cold Spring Harbor Laboratory Press; all rights reserved

Das könnte Ihnen auch gefallen