ArticlePDF Available

Abstract and Figures

In this review, most of the known and postulated mechanisms of osteopontin (OPN) and its role in bone remodeling and orthodontic tooth movement are discussed based on available literature. OPN, a multifunctional protein, is considered crucial for bone remodeling, biomineralization, and periodontal remodeling during mechanical tension and stress (orthodontic tooth movement). It contributes to bone remodeling by promoting osteoclastogenesis and osteoclast activity through CD44- and αvβ3-mediated cell signaling. Further, it has a definitive role in bone remodeling by the formation of podosomes, osteoclast survival, and osteoclast motility. OPN has been shown to have a regulatory effect on hydroxyapatite crystal (HAP) growth and potently inhibits the mineralization of osteoblast cultures in a phosphate-dependent manner. Bone remodeling is vital for orthodontic tooth movement. Significant compressive and tensional forces on the periodontium induce the signaling pathways mediated by various osteogenic genes including OPN, bone sialoprotein, Osterix, and osteocalcin. The signaling pathways involved in the regulation of OPN and its effect on the periodontal tissues during orthodontic tooth movement are further discussed in this review. A limited number of studies have suggested the use of OPN as a biomarker to assess orthodontic treatment. Furthermore, the association of single nucleotide polymorphisms (SNPs) in OPN coding gene Spp1 with orthodontically induced root resorption remains largely unexplored. Accordingly, future research directions for OPN are outlined in this review.
Content may be subject to copyright.
R E V I E W Open Access
Role of osteopontin in bone remodeling
and orthodontic tooth movement: a review
Amarjot Singh
1*
, Gurveen Gill
1,3
, Harsimrat Kaur
1,3
, Mohamed Amhmed
1,3
and Harpal Jakhu
2,4
Abstract
In this review, most of the known and postulated mechanisms of osteopontin (OPN) and its role in bone
remodeling and orthodontic tooth movement are discussed based on available literature. OPN, a multifunctional
protein, is considered crucial for bone remodeling, biomineralization, and periodontal remodeling during
mechanical tension and stress (orthodontic tooth movement). It contributes to bone remodeling by promoting
osteoclastogenesis and osteoclast activity through CD44- and αvβ3-mediated cell signaling. Further, it has a
definitive role in bone remodeling by the formation of podosomes, osteoclast survival, and osteoclast motility.
OPN has been shown to have a regulatory effect on hydroxyapatite crystal (HAP) growth and potently inhibits the
mineralization of osteoblast cultures in a phosphate-dependent manner. Bone remodeling is vital for orthodontic
tooth movement. Significant compressive and tensional forces on the periodontium induce the signaling pathways
mediated by various osteogenic genes including OPN, bone sialoprotein, Osterix, and osteocalcin. The signaling
pathways involved in the regulation of OPN and its effect on the periodontal tissues during orthodontic tooth
movement are further discussed in this review. A limited number of studies have suggested the use of OPN as a
biomarker to assess orthodontic treatment. Furthermore, the association of single nucleotide polymorphisms (SNPs)
in OPN coding gene Spp1 with orthodontically induced root resorption remains largely unexplored. Accordingly,
future research directions for OPN are outlined in this review.
Keywords: Osteopontin, Bone remodeling, Biomarkers, Root resorption, Orthodontic tooth movement
Background
Osteopontin (OPN) is a highly phosphorylated and gly-
cosylated sialoprotein that is expressed by several cell
types including osteoblasts, osteocytes, and odontoblasts.
OPN belongs to the family of non-collagenous proteins
known as SIBLING (small integrin-binding ligand,
N-linked glycoprotein) [1]. In humans, OPN is encoded
by Spp1 gene located on the long arm of chromosome 4
region 22 (4q1322.1). OPN is a prominent component of
mineralized extracellular matrices of bones and teeth
[2]. It has been found to be involved in a number of
pathologic and physiological events including bone re-
modeling, biomineralization, wound healing, apoptosis,
and tumor metastasis [2].
Bone remodeling is crucial for maintaining the normal
skeletal structure as well as a key factor for orthodontic
tooth movement. Orthodontic forces exert a significant
amount of compressive [39] and tensional [7,1013]
forces on the periodontium to induce the signaling path-
ways mediated by various osteogenic genes including
OPN, bone sialoprotein, Osterix, and osteocalcin. The
signaling pathways and response of the periodontium
differ on both tension and compression sides; however,
OPN is ubiquitously expressed in bone remodeling on
both sides [13].
In this review, our focus will be on the events con-
trolled by OPN in bone remodeling and orthodontic
tooth movement. In addition, the prospects of OPN in
accelerating tooth movement and root resorption and as
a biomarker will be outlined. In our knowledge, no study
till date has reviewed the mechanisms involved in
OPN-mediated bone remodeling during orthodontic
tooth movement.
OPN structure and its expression and regulation
OPN is multifunctional protein owing to its structure.
OPN molecule comprises unique conserved regions
* Correspondence: amarjot.singh@mail.mcgill.ca
1
Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
Full list of author information is available at the end of the article
© The Author(s). 2018 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and
reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license, and indicate if changes were made.
Singh et al. Progress in Orthodontics (2018) 19:18
https://doi.org/10.1186/s40510-018-0216-2
which involve (RG)-binding domain, serine/threonine
phosphorylation site, two heparin-binding sites, one
thrombin cleavage site, and a putative calcium-
binding site [14]. The cell interacting domains include
arginine-glycine-aspartic acid (RGD) cell-binding se-
quence and serine-valine-valine-tyrosine-glutamate-
leucine-arginine (SVVYGLR) motif [15]. The cleavage
sites include thrombin and matrix metalloprotinases
(MMPs) cleavage sites [14]. In response to cleavage
by thrombin, SVVYGLR site is revealed and leads to the
formation of two segments: N-terminal fragment and
C-terminal fragment (Fig. 1). The pro-inflammatory
N-terminal segment includes two integrin-binding sites:
RGD and SVVYGLR motifs [15]. However, the C-terminal
fragment is devoid of an integrin-binding site. MMPs
cleave both fragments by binding to MMPs cleavage sites:
cleaving N-terminal fragment leads to inactivation of
integrin-binding domain of SVVYGLR motifs [15].
The expression of OPN is regulated by a large number
of cytokines, hormones, and growth factors, which affects
gene transcription, translation, and post-translational
modifications (Table 1)[16]. Also, expression of OPN in-
creases in response to mechanical stress [1719]. There-
fore, it is a critical factor in regulating bone remodeling in
responses to mechanical stimuli.
OPN in bone remodeling
OPN is considered to play important role in bone forma-
tion and resorption [2022]. It is highly concentrated at
cement lines where pre-existing and newly formed bone
meet and at bone surfaces interfacing with cells called as
laminae limitantes [23]. There are various levels of medi-
ation of OPN in bone remodeling. For example, OPN is
demonstrated to have chemotactic activity [24]onthe
precursor of osteoclasts, at a concentration from 10 nM
to 1 μM[17]. Also, OPN-dependent intracellular signaling
is seen in sealing zone formation in osteoclastic resorption
(Fig. 2a,b). Broadly, various authors described the follow-
ing pathways in OPN-mediated bone remodeling.
Integrin αvβ3-mediated signaling
OPN binds to several integrins including αvβ3, αvβ5,
αvβ1, α4β1, α5, and α9β1. OPN binding to αvβ3iscrucial
for major post-receptor signal responses, which involves
regulation of osteoclastic activity and activation of osteo-
protegerin expression [24,25]. Further, OPN binding to
integrin αvβ3 plays a major role in the formation of
sealing zone in osteoclast activity. OPN-αvβ3bindingon
the surface of osteoclasts induces integrin clustering and
leads to intracellular signaling by phosphorylation of pro-
tein tyrosine kinase 2 (PYK2) [25,26]thatfacilitatebind-
ing of proto-oncogene tyrosine-protein kinase (Src) via its
SH2 domain. This Src-PYK2 binding leads to further
phosphorylation of PYK2 at other sites which amplifies
the signals activating cellular functions including cell
adhesion such as sealing zone formation (Fig. 2b)[25,26].
It has also been suggested that integrin αvβ3, Src, and Fms
(the receptor for M-CSF) stimulate Spleen tyrosine kinase
(Syk) which further mediates GTP loading on Rac1 via Vav3
in osteoclasts [27]. GTP loading on Rac 1 drives cytoskeletal
remodeling leading to bone resorption. Certain proteins
including Wiskott-Aldrich syndrome protein (WASP) and
gelsolin are also regulated by integrin αvβ3. This process is
vital for the podosome formation on osteoclasts [27].
In addition, OPN binding to integrin αvβ3 has been sug-
gested to modulate intracellular Ca
2+
through stimulation
of Ca
2+
release from intracellular compartments and regu-
lating extracellular calcium influx via Ca
2+
-ATPase pump
[28,29]. The induction of cytosolic Ca
2+
further modu-
lates osteoclast activity by translocation of transcription
factor NFATc1 (nuclear factor of activated T cells,
cytoplasmic 1) through the Ca
2+
-NFAT pathway (Fig. 2b)
[30,31]. This NFATc1 has been shown to be imperative
for osteoclastogenesis [3234], leading to the increased re-
sorptive activity of mature osteoclasts [30,31].
CD44-associated cell signaling
Osteoclasts deficient in OPN show no migratory activity
and do not resorb bone [35]. It has been demonstrated
Fig. 1 A schematic representation of osteopontin structure and thrombin cleavage site. RGD (arginine-glycine-aspartic acid) and SVVYGLR
(serine-valine-valine-tyrosine-glutamate-leucine-arginine) binding domains are indicated
Singh et al. Progress in Orthodontics (2018) 19:18 Page 2 of 8
that OPN-deficient osteoclasts, when treated with
exogenous OPN, result in an enhanced CD44 expres-
sion [36]. CD44-induced cell signaling enhances
osteoclast motility [35], which partially restores bone
resorption, by activation of αvβ3integrin[36,37].
OPN stimulate osteoclast migration through αvβ3-
and CD44-mediated cell signaling, which further in-
creases CD44 expression on osteoclasts [35,36].
Addition of exogenous OPN partially restores the
resorptive activity of osteoclasts, which indicates
autocrine OPN is important to osteoclast activity
[36]. However, exogenously added OPN does not
have access to OPN secreted by osteoclasts, which
are present in resorption lacuna [36]. The intracellu-
lar form of OPN (iOPN), an integral component of
the CD44-ERM complex, is seen to be involved in
migrating fibroblasts, macrophages, osteoclasts, and
metastatic breast cancer lines [2,38]. A hypothetical
pathway was described in which iOPN with components
of CD44-ERM is involved in cell migration [2,38].
Further, it has been demonstrated that overexpression of
phosphatase and tensin homolog (PTEN) restricts
PI3-kinase signaling, suppresses receptor activator of
nuclear kappa-B ligand (RANKL) and OPN-induced Akt
activation, and ultimately results in the downregulation of
osteoclast differentiation and cell motility [39].
Table 1 Factors affecting the expression and regulation of osteopontin
Expression and upregulation of OPN Downregulation of OPN
Transcription factorsRunx2 and Osterix [68] cGMP-dependent protein kinase [2]
Inorganic phosphate [69] Bisphosphonates [2]
Systematic conditionshypophosphatemia, hypocalcemia [2]
Hormonesglucocorticoids, [70]
1,25-dihydroxyvitamin D3, [70] parathyroid hormone [14]
ERK inhibitor
Vitaminsretinoic acid [70]
Inflammatory mediatorsTNFα, IL-1β, TGFβ[14]
Mechanical stress
ab
Fig. 2 A schematic representation of bone resorption occurring at cellular and molecular level. aRANKL/RANK/OPG pathway and osteopontin in
podosome formation. bOsteopontin binding to integrin αvβ3 leads to podosome formation and osteoclastic activity via Rac and NFAT pathway
respectively. M-CSF (macrophage colony-stimulating factor), CSF-R (colony-stimulating factor receptor), RANKL (receptor activator of nuclear
kappa-B ligand), RANK (receptor activator of nuclear kappa-B), OPG (osteoprotegerin), Src (proto-oncogene tyrosine-protein kinase), Syk (Spleen
tyrosine kinase), Vav3 (vav guanine nucleotide exchange factor 3), Rac1 (Ras-related C3 botulinum toxin substrate 1), NFAT (nuclear factor of
activated T cells)
Singh et al. Progress in Orthodontics (2018) 19:18 Page 3 of 8
Inhibition of mineral deposition
The bone matrix consists of the inorganic component, hy-
droxyapatite (HA), and organic component, proteins and
proteoglycans [2]. OPN protein along with other SIBLING
proteins contain acidic, serine-, and aspartate-rich motif
(ASARM) which are the potential phosphorylation sites
[1]. Phosphorylated OPN inhibits mineralization via phos-
phate residues [40]. Contrary to it, OPN dephosphoryla-
tion by tissue-non-specific alkaline phosphatase (TNAP)
prevents much of its mineral binding and crystal growth
activity [40]. Both pyrophosphate (PPi) and OPN contains
highly negative charge phosphate residues which inhibit
mineralization after binding to HA crystals [40]. It has
been shown that peptide phosphorylated MEPE ASARM
(pASARM) has a greater affinity for HA than nonpho-
sphorylated ASARM (npASARM). OPN can act inde-
pendently of PPi as well as a mediator of PPi effects. High
levels of extracellular PPi lead to increased OPN expres-
sion and secretion by osteoblasts [40].
Pyrophosphate prevents mineralization by three pro-
posed mechanisms. Firstly, there is direct binding of PPi
to growing HA crystals. Secondly, there is the induction
of OPN expression by osteoblasts through MAPK path-
way, enabling the coordinated action of both PPi and
OPN [40]. Thirdly, there is a feedback mechanism in
which Pi/PPi ratio inhibits TNAP activity [40]. Even
though OPN is considered as mineralization inhibitor, it
has been shown that OPN can serve as an agent for
intra-fibrillar mineralization in collagen [41], thus pointing
towards the multifunctional role of OPN.
Potential role of OPN in orthodontic tooth
movement
Various knockout studies have demonstrated that bone
remodeling is impaired in OPN-deficient mice [42]in
response to mechanical stress [8,43,44]. An animal
study [44], by Walker et al., has revealed that OPN is
required for osteoclast recruitment through RANKL
expression in unloaded mechanical stress (unopposed
molar model). Further, it has been suggested that OPN
mediates osteoclast activity, RANKL expression, and
bone resorption at unloaded alveolar bone walls using a
PI3K- and ERK-dependent mechanism [44]. No distal
drifting was reported in the OPN-deficient mice [44].
In the initial stages of orthodontic tooth movement,
OPN is observed in the osteocytes [13]. A study [17]
suggested the change in the number of OPN mRNA
expressing osteocytes on the pressure side after 48 h of
mechanical stress and reached a maximum value at 72 h
[8], coinciding with bone resorption. However, in the
later stages of OTM, OPN is ubiquitously expressed in
PDL cells, osteoclasts, cementocytes, cementoblasts, and
osteoblasts as well as the cement line of alveolar bone
and cementum [13,45,46]. The potential signaling
pathways involved in the OPN regulation during the
orthodontic tooth movement on compression as well as
on tension side are summarized in Fig. 3.
OPN and RANKL regulation on compression side
Wongkhantee and coworkers first studied the OPN ex-
pression in human periodontal ligament cell (HPDL) via
Rho kinase pathway (Fig. 3)[4] and analyzed that
stress-induced ATP activates Rho kinase pathway via the
purinoreceptor 1 (P2Y1) receptor [5]. They proposed
that RANKL upregulation during mechanical compres-
sion may be further induced via activation of NFκB
pathway-mediated release of cyclooxygenase and prosta-
glandin E2 (PGE2) production [3]. Later, various re-
search groups analyzed the Rho kinase-mediated OPN
induction. Hong et al. reported that OPN induction dur-
ing compression is mediated by RhoA-controlled focal
adhesion kinase (FAK) and extracellular signal-regulated
kinase (ERK) pathways in human periodontal ligament
fibroblasts (Fig. 3). ERK further phosphorylates ETS
domain-containing protein (Elk-1) which results in the
transcription of OPN [47].
OPN and RANKL collectively work to induce the bone
resorption in response to compressive forces (Fig. 3).
Osteoblasts and stromal stem cells express receptor
activator RANKL which binds to its receptor, receptor
activator of nuclear kappa-B (RANK), on the surface of
osteoclasts and their precursors. This regulates the
differentiation of precursors into multinucleated osteo-
clasts [48,49]. In addition, a study by Walker and
coworkers suggested that increased OPN expression
enhances RANKL expression via extracellular matrix sig-
naling pathway in unloaded distal drift [44]. Nevertheless,
no study has assessed the influence of OPN expression on
RANKL in mechanically stressed condition viz. orthodon-
tic tooth movement and need further investigation.
OPN regulation on tension side
Su et al. first reported the expression of a gap junction
alpha-1 protein, connexin 43, on tension side during
orthodontic tooth movement in rat periodontal ligament
cells [10]. Later, Shengnan et al. confirmed the involve-
ment of connexin 43 and ERK in tension-induced signal
transduction human periodontal ligament fibroblasts
(Fig. 3)[7]. It was reported that ERK further induces the
transcription of osteogenic proteins, runt-related tran-
scription factor 2 (RUNX2), osteoprotegerin (OPG), and
Osterix [7]. In a recent study, the upregulation of OPN
along with alkaline phosphatase, collagen I, osteocalcin,
and bone sialoprotein was reported via ERK and p38
MAPK-mediated pathway during orthodontic tooth
movement in response to tension stress [11]. Thus, both
ERK and p38 were proposed to be significantly involved
Singh et al. Progress in Orthodontics (2018) 19:18 Page 4 of 8
in periodontal remodeling during orthodontic tooth
movement [11].
Wnt/βcatenin pathway has been shown to be signifi-
cantly involved in the matrix formation in response to
mechanical strain [5054]. Whether this pathway is in-
volved in the tension forces created during the ortho-
dontic tooth movement is not yet known. Thus, we
hypothesize that strain-induced transduction of Wnt/β
catenin could be involved in the upregulation of osteo-
genic proteins including Osterix and OPN (Fig. 3).
OPN-mediated tooth root resorption and repair
Root resorption is one of the side effects of the ortho-
dontic treatment and is the result of activity of odonto-
clasts [45]. A mice study showed odontoclast expressing
OPN mRNA appeared on the surface of the active root
resorption 5 days after orthodontic movement [45].
Similarly, Chung et al. demonstrated that OPN defi-
ciency has much more enhanced effect on the decrease
in the odontoclastic activity than osteoclastic activity
[43]. They proposed that abundance of inflammatory
regulators in the alveolar bone might overwhelm the
deficiency of OPN, thereby having little effect on the bone
resorption [43]. In contrast to the alveolar bone, cementum
and root surface of the tooth is deficient in the inflamma-
tory mediators, thereby enhanced odontoclastic activity
may be the one reason in OPN-deficient mice [43]. Thus,
OPN is a crucial factor in force-induced root resorption of
tooth [43]. Jimenez-Pellegrin et al. demonstrated that OPN
plays a key role in both cementum resorption and repair
after orthodontic rotation movement [55].
On the other hand, the role of OPN in cementogenesis
followed by mechanical injury was also studied in the
epithelial cell rests of Malassez (ECRM) [56,57]. It has
been suggested that ECRM express various osteogenic
genes including OPG and OPN [56]. Also, immunohis-
tochemical characteristics of ECRM suggested that it
may be significantly involved in the secretion of matrix
proteins including OPN to further induce cementum
repair followed by mechanical injury [57].
Various research groups studied the single nucleotide
polymorphisms (SNPs) in the OPN coding gene Spp1
and its effect on the tooth root resorption [5860].
Iglesias-Linares and coworkers first reported that OPN
Fig. 3 A schematic representation of osteopontin regulation and osteopontin-mediated periodontal remodeling during orthodontic tooth movement
at tension side and compression side. ECM (extra-cellular matrix), PDL (periodontal ligament), Cx43 (connexin 43), ERK1/2 (extra-cellular signal-regulated
kinase 1,2), RUNX2 (runt-related transcription factor 2), IL-1/IL-8 (interleukin 1/8), MMPs (matrix metalloproteinases), VEGF (vascular endothelial growth
factor), TIMPs (tissue inhibitors of metalloproteinases), ATP (adenosine triphosphate), PGE2 (prostaglandin E2), EP, RANKL (receptor activator of nuclear
kappa-B ligand), RANK (receptor activator of nuclear kappa-B), OPG (osteoprotegerin), P2Y1 (purinoreceptor 1), Pka (protein kinase A), NFkB (nuclear
factor kappa B), COX (cyclooxygenase), ROCK (Rho-associated protein kinase), FAK (focal adhesion kinase), ELK1 (ETS domain containing protein), AP1
(activator protein 1)
Singh et al. Progress in Orthodontics (2018) 19:18 Page 5 of 8
gene SNPs (rs9138, rs11730582) are involved in the sus-
ceptibility of external root resorption in patients under-
going orthodontic treatment [58]. However, in another
study, OPN gene SNPs and its effect on external apical
root resorption (EARR) were not confirmed in Czech
children [60]. However, the association between individual
variability in purinoreceptor (P2X7) and EARR was sug-
gested to be an important factor in the etiopathogenesis of
EARR [60]. Iglesias-Linares et al. later implicated the Spp1
gene SNPs to assess the orthodontically induced external
apical root resorption (OIEARR) in patients with remov-
able appliances versus fixed appliances [59]. No any pre-
disposition to OIEARR was reported with response to
fixed and removable appliances [59].
Future directions
Since OPN is ubiquitously expressed in periodontal re-
modeling during orthodontic tooth movement, various
research groups have implicated OPN as a biomarker to
assess the tissue response with respect to orthodontic
treatment [61]. The samples were collected from GCF
and a protein levels were assessed [6165]. DNA methy-
lation biomarkers of Spp1 gene and other osteogenic
genes may also be helpful to understand the individual
variability in response to orthodontic treatment [66].
Thus, a more tailored and personalized approach [66]
can be drawn to treat patients with an increased predis-
position to OIEARR via targeting the epigenetic mecha-
nisms. Similarly, micro RNAs targeting the osteogenic
genes can be assessed.
Alveolar decortication has been shown to induce the
rate of tooth movement via the coupled mechanism of
bone resorption and formation in early stages of ortho-
dontic tooth movement [67]. The underlying biomarkers
(OPN, osteocalcin, bone sialoprotein) demonstrated in-
creased anabolic activity. Whether the orthodontic tooth
movement can be accelerated via targeting the under-
lying signaling pathways warrants further investigation.
Conclusions
OPN has a definitive role in the formation of podo-
somes, osteoclast survival, and osteoclast motility. Vari-
ous OPN-mediated signaling pathways involved in the
periodontal remodeling facilitate orthodontic tooth
movement. There is a need to pharmacologically target
these signaling pathways in order to decrease the side ef-
fects of orthodontic treatment including tooth root re-
sorption in patients with an increased predisposition to
OIEARR. In addition, the application of OPN bio-
markers should be assessed and compared at proteomic,
genomic, and epigenomic levels in order to gain a more
tailored orthodontic approach. Nonetheless, there is dire
need of validated studies to further translate the rele-
vance of OPN in orthodontic treatment.
Abbreviations
AP1: Activator protein 1; ATP: Adenosine triphosphate; COX: Cyclooxygenase;
CSF-R: Colony-stimulating factor receptor; Cx43: Connexin 43;
ECM: Extracellular matrix; ELK1: ETS domain containing protein; EP,
P2Y1: Purinoreceptor 1; ERK1/2: Extracellular signal-regulated kinase 1,2;
FAK: Focal adhesion kinase; IL-1/IL-8: Interleukin 1/8; M-CSF: Macrophage
colony-stimulating factor; MMPs: Matrix metalloproteinases; NFAT: Nuclear
factor of activated T cells; NFkB: Nuclear factor kappa B;
OPG: Osteoprotegerin; PDL: Periodontal ligament; PGE2: Prostaglandin E2;
Pka: Protein kinase A; Rac1: Ras-related C3 botulinum toxin substrate 1;
RANK: Receptor activator of nuclear kappa-B; RANKL: Receptor activator of
nuclear kappa-B ligand; RGD: Arginine-glycine-aspartic acid; ROCK: Rho-
associated protein kinase; RUNX2: Runt related transcription factor 2;
Src: Proto-oncogene tyrosine-protein kinase; SVVYGLR: Serine-valine-valine-
tyrosine-glutamate-leucine-arginine; Syk: Spleen tyrosine kinase; TIMPs: Tissue
inhibitors of metalloproteinases; Vav3: Vav guanine nucleotide exchange
factor 3; VEGF: Vascular endothelial growth factor
Authorscontributions
AS made a substantial contribution to the conception, design, and revision
of the manuscript. GG, HK, MA, and HJ made contributions in the revision of
the manuscript. AS and HK designed the figures. All authors read and
approved the final manuscript.
Ethics approval and consent to participate
Ethical approval was not required.
Competing interests
The authors declare that they have no competing interests.
PublishersNote
Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional affiliations.
Author details
1
Faculty of Dentistry, McGill University, Montreal, Quebec, Canada.
2
Department of Endodontics, Government Dental College, Amritsar, Punjab,
India.
3
Lady Davis Institute, Jewish General Hospital, Montreal, Quebec,
Canada.
4
Sandalwood Smiles, Private Dental Practice, Brampton, Ontario,
Canada.
Received: 27 February 2018 Accepted: 24 May 2018
References
1. Staines KA, MacRae VE, Farquharson C. The importance of the SIBLING
family of proteins on skeletal mineralisation and bone remodelling.
J Endocrinol. 2012;214:24155.
2. Sodek J, Ganss B, McKee MD. Osteopo ntin. Crit Rev Oral Biol Med.
2000;11:279303.
3. Luckprom P, Wongkhantee S, Yongchaitrakul T, Pavasant P. Adenosine
triphosphate stimulates RANKL expression through P2Y 1receptor-cyclo-
oxygenase-dependent pathway in human periodontal ligament cells.
J Periodontal Res. 2010;45:40411.
4. Wongkhantee S, Yongchaitrakul T, Pavasant P. Mechanical stress induces
osteopontin expression in human periodontal ligament cells through rho
kinase. J Periodontol. 2007;78:11139.
5. Wongkhantee S, Yongchaitrakul T, Pavasant P. Mechanical stress induces
osteopontin via ATP/P2Y1 in periodontal cells. J Dent Res. 2008;87:5648.
6. Luckprom P, Kanjanamekanant K, Pavasant P. Role of connexin43
hemichannels in mechanical stress-induced ATP release in human
periodontal ligament cells. J Periodontal Res. 2011;46:60715.
7. Li S, Zhang H, Li S, Yang Y, Huo B, Zhang D. Connexin 43 and ERK regulate
tension-induced signal transduction in human periodontal ligament
fibroblasts. J Orthop Res. 2015;33:100814.
8. Fujihara S, Yokozeki M, Oba Y, Higashibata Y, Nomura S, Moriyama K.
Function and regulation of osteopontin in response to mechanical stress.
J Bone Miner Res. 2006;21:95664.
9. Mitsui N, Suzuki N, Maeno M, Mayahara K, Yanagisawa M, Otsuka K, Shimizu
N. Optimal compressive force induces bone formation via increasing bone
Singh et al. Progress in Orthodontics (2018) 19:18 Page 6 of 8
sialoprotein and prostaglandin E2 production appropriately. Life Sci. 2005;
77:316882.
10. Su M. Expression of connexin 43 in rat mandibular bone and periodontal
ligament (PDL) cells during experimental tooth movement. J Dent Res.
1997;76:135766.
11. Jiang L, Tang Z. Expression and regulation of the ERK1/2 and p38 MAPK
signaling pathways in periodontal tissue remodeling of orthodontic tooth
movement. Mol Med Rep. 2017;17:1499506.
12. Huang L, Meng Y, Ren A, Han X, Bai D, Bao L. Response of cementoblast-
like cells to mechanical tensile or compressive stress at physiological levels
in vitro. Mol Biol Rep. 2009;36:17418.
13. Kim JY, Kim BI, Jue SS, Park JH, Shin JW. Localization of osteopontin and
osterix in periodontal tissue during orthodontic tooth movement in rats.
Angle Orthod. 2012;82:10714.
14. Kahles F, Findeisen HM, Bruemmer D. Osteopontin: a novel regulator at the
cross roads of inflammation, obesity and diabetes. Mol Metab. 2014;3:38493.
15. Wolak T. Osteopontina multi-modal marker and mediator in
atherosclerotic vascular disease. Atherosclerosis. 2014;236:32737.
16. Ishii T, Ohshima S, Ishida T, et al. Osteopontin as a positive regulator in the
osteoclastogenesis of arthritis. Biochem Biophys Res Commun. 2004;316:80915.
17. Terai K, Takano-Yamamoto T, Ohba Y, Hiura K, Sugimoto M, Sato M, Kawahata
H, Inaguma N, Kitamura Y, Nomura S. Role of osteopontin in bone remodeling
caused by mechanical stress. J Bone Miner Res. 1999;14:83949.
18. Morinobu M, Ishijima M, Rittling SR, Tsuji K, Yamamoto H, Nifuji A, Denhardt
DT, Noda M. Osteopontin expression in osteoblasts and osteocytes during
bone formation under mechanical stress in the calvarial suture in vivo.
J Bone Miner Res. 2003;18:170615.
19. Jeon YM, Kook SH, Son YO, Kim EM, Park SS, Kim JG, Lee JC. Role of MAPK in
mechanical force-induced up-regulation of type I collagen and osteopontin in
human gingival fibroblasts. Mol Cell Biochem. 2009;320:4552.
20. Denhardt DT, Guo X. Osteopontin: a protein with diverse functions. FASEB J.
1993;7:147582.
21. Butler WT. The nature and significance of osteopontin. Connect Tissue Res.
1989;23:12336.
22. McKee MD, Farach-Carson MC, Butler WT, Hauschka PV, Nanci A.
Ultrastructural immunolocalization of noncollagenous (osteopontin and
osteocalcin) and plasma (albumin and α2HS-glycoprotein) proteins in rat
bone. J Bone Miner Res. 1993;8:48596.
23. McKee MD, Nanci A. Osteopontin and the bone remodeling sequence:
colloidal-gold immunocytochemistry of an interfacial extracellular matrix
protein. Ann N Y Acad Sci. 1995;760:17789.
24. Standal T, Borset M, Sundan A. Role of osteopontin in adhesion, migration,
cell survival and bone remodeling. Exp Oncol. 2004;26:17984.
25. Duong LT, Lakkakorpi P, Nakamura I, Rodan GA. Integrins and signaling in
osteoclast function. Matrix Biol. 2000;19:97105.
26. Duong LT, Rodan GA. Integrin-mediated signaling in the regulation of
osteoclast adhesion and activation. Front Biosci. 1998;3:75768.
27. Huveneers S, Danen EHJ. Adhesion signalingcrosstalk between integrins,
Src and rho. J Cell Sci. 2009;122:105969.
28. Zimolo Z, Wesolowski G, Tanaka H, Hyman JL, Hoyer JR, Rodan GA. Soluble
alpha v beta 3-integrin ligands raise [Ca2+]i in rat osteoclasts and mouse-
derived osteoclast-like cells. Am J Phys. 1994;266:C37681.
29. Miyauchi A, Alvarez J, Greenfield EM, et al. Recognition of osteopontin and
related peptides by an α(v)β3 integrin stimulates immediate cell signals in
osteoclasts. J Biol Chem. 1991;266:2036974.
30. Tanabe N, Wheal BD, Kwon J, Chen HH, Shugg RPP, Sims SM, Goldberg HA,
Dixon SJ. Osteopontin signals through calcium and nuclear factor of
activated T cells (NFAT) in osteoclasts: a novel RGD-dependent pathway
promoting cell survival. J Biol Chem. 2011;286:3987181.
31. Kikuta J, Ishii M. Osteoclast migration, differentiation and function: novel
therapeutic targets for rheumatic diseases. Rheumatology. 2013;52:22634.
32. Negishi-Koga T, Takayanagi H. Ca2+-NFATc1 signaling is an essential axis of
osteoclast differentiation. Immunol Rev. 2009;231:24156.
33. Komarova SV, Pereverzev A, Shum JW, Sims SM, Dixon SJ. Convergent
signaling by acidosis and receptor activator of NF-kappaB ligand (RANKL)
on the calcium/calcineurin/NFAT pathway in osteoclasts. Proc Natl Acad Sci
U S A. 2005;102:26438.
34. Ikeda F, Nishimura R, Matsubara T, Hata K, Reddy SV, Yoneda T.
Activation of NFAT signal in vivo leads to osteo penia associated with
increased osteoclastogenesis and bone-resorbing activity. J Immunol.
2006;177:238490.
35. Chellaiah MA, Hruska KA. The integrin {alpha}
v
{beta}
3
andCD44regulatethe
actions of osteopontin on osteoclast motility. Calcif Tissue Int. 2003;72:197205.
36. Chellaiah MA. Osteopontin deficiency produces osteoclast dysfunction due
to reduced CD44 surface expression. Mol Biol Cell. 2003;14:17389.
37. Rangaswami H, Bulbule A, Kundu GC. Osteopontin: role in cell signaling and
cancer progression. Trends Cell Biol. 2006;16:7987.
38. Zohar R, Suzuki N, Suzuki K, Arora P, Glogauer M, Mcculloch CAG, Sodek J.
Intracellular osteopontin is an integral component of the CD44-ERM
complex involved in cell migration. J Cell Physiol. 2000;184:11830.
39. Sugatani T, Alvarez U, Hruska KA. PTEN regulates RANKL- and osteopontin-
stimulated signal transduction during osteoclast differentiation and cell
motility. J Biol Chem. 2003;278:50018.
40. Sapir-Koren R, Livshits G. Bone mineralization and regulation of phosphate
homeostasis. IBMS Bonekey. 2011;8:286300.
41. Rodriguez DE, Thula-Mata T, Toro EJ, Yeh YW, Holt C, Holliday LS, Gower LB.
Multifunctional role of osteopontin in directing intrafibrillar mineralization of
collagen and activation of osteoclasts. Acta Biomater. 2014;10:494507.
42. Foster BL, Ao M, Salmon CR, et al. Osteopontin regulates dentin and
alveolar bone development and mineralization. Bone. 2018;107:196207.
43. Chung CJ, Soma K, Rittling SR, Denhardt DT, Hayata T, Nakashima K, Ezura Y,
Noda M. OPN deficiency suppresses appearance of odontoclastic cells and
resorption of the tooth root induced by experimental force application. J
Cell Physiol. 2008;214:61420.
44. Walker CG, Dangaria S, Ito Y, Luan X, Diekwisch TGH. Osteopontin is
required for unloading-induced osteoclast recruitment and modulation of
RANKL expression during tooth drift-associated bone remodeling, but not
for super-eruption. Bone. 2010;47:10209.
45. Kuroda S, Balam TA, Sakai Y, Tamamura N, Takano-Yamamoto T. Expression
of osteopontin mRNA in odontoclasts revealed by in situ hybridization
during experimental tooth movement in mice. J Bone Miner Metab. 2005;
23:1103.
46. Takano-Yamamoto T, Takemura T, Kitamura Y, Nomura S. Site-specific
expression of mRNAs for osteonectin, osteocalcin, and osteopontin revealed
by in situ hybridization in rat periodontal ligament during physiological
tooth movement. J Histochem Cytochem. 1994;42:88596.
47. Hong SY, Jeon YM, Lee HJ, Kim JG, Baek JA, Lee JC. Activation of RhoA and
FAK induces ERK-mediated osteopontin expression in mechanical force-
subjected periodontal ligament fibroblasts. Mol Cell Biochem. 2010;335:26372.
48. Boyce BF, Xing L. The RANKL/RANK/OPG pathway. Curr Osteoporos Rep.
2007;5:98104.
49. Udagawa N, Takahashi N, Jimi E, et al. Osteoblasts/stromal cells stimulate
osteoclast activation through expression of osteoclast differentiation factor/
RANKL but not macrophage colony-stimulating factor. Bone. 1999;25:51723.
50. Han P, Ivanovski S, Crawford R, Xiao Y. Activation of the canonical Wnt
signaling pathway induces cementum regeneration. J Bone Miner Res. 2015;
30:116074.
51. Lim WH, Liu B, Cheng D, Williams BO, Mah SJ, Helms JA. Wnt signaling
regulates homeostasis of the periodontal ligament. J Periodontal Res. 2014;
49:7519.
52. Lim WH, Liu B, Mah S, Yin X, Helms JA. Alveolar bone turnover and
periodontal ligament width are controlled by Wnt. J Periodontol. 2015;
86:31926.
53. Holguin N, Brodt MD, Silva MJ. Activation of Wnt signaling by
mechanical loading is impaired in the bone of old mice. J Bone Miner
Res. 2016;31 :221526.
54. Tu X, Rhee Y, Condon KW, et al. Sost downregulation and local Wnt
signaling are required for the osteogenic response to mechanical loading.
Bone. 2012;50:20917.
55. Jimenez-Pellegrin C, Arana-Chavez VE. Root resorption repair in mandibular
first premolars after rotation. A transmission electron microscopy analysis
combined with immunolabeling of osteopontin. Am J Orthod Dentofac
Orthop. 2007;132:2306.
56. Mizuno N, Shiba H, Mouri Y, Xu W, Kudoh S, Kawaguchi H, Kurihara H.
Characterization of epithelial cells derived from periodontal ligament by
gene expression patterns of bone-related and enamel proteins. Cell Biol Int.
2005;29:1117.
57. Hasegawa N, Kawaguchi H, Ogawa T, Uchida T, Kurihara H.
Immunohistochemical characteristics of epithelial cell rests of Malassez
during cementum repair. J Periodontal Res. 2003;38:516.
58. Iglesias-Linares A, Yañez-Vico RM, Moreno-Fernández AM, Mendoza-
Mendoza A, Orce-Romero A, Solano-Reina E. Osteopontin gene SNPs
Singh et al. Progress in Orthodontics (2018) 19:18 Page 7 of 8
(rs9138, rs11730582) mediate susceptibility to external root resorption in
orthodontic patients. Oral Dis. 2014;20:30712.
59. Iglesias-Linares A, Sonnenberg B, Solano B, Yañez-Vico RM, Solano E,
Lindauer SJ, Flores-Mir C. Orthodontically induced external apical root
resorption in patients treated with fixed appliances vs removable aligners.
Angle Orthod. 2017;87:310.
60. Borilova Linhartova P, Cernochova P, Kastovsky J, Vrankova Z, Sirotkova M,
Izakovicova Holla L. Genetic determinants and postorthodontic external
apical root resorption in Czech children. Oral Dis. 2017;23:2935.
61. Castroflorio T, Gamerro EF, Caviglia GP, Deregibus A. Biochemical markers of
bone metabolism during early orthodontic tooth movement with aligners.
Angle Orthod. 2017;87:7481.
62. Barbieri G, Solano P, Alarcón JA, Vernal R, Rios-Lugo J, Sanz M, Martín C.
Biochemical markers of bone metabolism in gingival crevicular fluid during
early orthodontic tooth movement. Angle Orthod. 2013;83:639.
63. George A, Evans CA. Detection of root resorption using dentin and bone
markers. Orthod Craniofacial Res. 2009;12:22935.
64. Kido J, Nakamura T, Asahara Y, Sawa T, Kohri K, Nagata T. Osteopontin in
gingival crevicular fluid. J Periodontal Res. 2001;36:32833.
65. Smuthkochorn S, Palomo JM, Hans MG, Jones CS, Palomo L. Gingival crevicular
fluid bone turnover biomarkers: how postmenopausal women respond to
orthodontic activation. Am J Orthod Dentofac Orthop. 2017;152:337.
66. Singh A, Gill G, Jakhu H, Gill G. Epigenetics and its implications for oral
health. J Oral Biosci. 2018; https://doi.org/10.1016/j.job.2018.02.001.
67. Baloul SS, Gerstenfeld LC, Morgan EF, Carvalho RS, Van Dyke TE, Kantarci A.
Mechanism of action and morphologic changes in the alveolar bone in
response to selective alveolar decortication-facilitated tooth movement. Am
J Orthod Dentofac Orthop. 2011;139:S83101.
68. Harada H, Tagashira S, Fujiwara M, Ogawa S, Katsumata T, Yamaguchi A,
Komori T, Nakatsuka M. Cbfa1 isoforms exert functional differences in
osteoblast differentiation. J Biol Chem. 1999;274:69728.
69. Fatherazi S, Matsa-Dunn D, Foster BL, Rutherford RB, Somerman MJ,
Presland RB. Phosphate regulates osteopontin gene transcription. J Dent
Res. 2009;88:3944.
70. Pike JW, Lee SM, Meyer MB. Regulation of gene expression by 1,25-
dihydroxyvitamin D3 in bone cells: exploiting new approaches and defining new
mechanisms. Bonekey Rep; 2014. https://doi.org/10.1038/bonekey.2013.216.
Singh et al. Progress in Orthodontics (2018) 19:18 Page 8 of 8
... 205 OPN plays an important role in bone remodeling. 206 High levels of blood OPN in postmenopausal women were associated with low BMD and osteoporotic vertebral fractures. 207,208 Clinical investigations demonstrated elevated levels of OPN in both the plasma and CSF of AD patients, [209][210][211] suggesting that OPN might play some role in the pathophysiology of AD. ...
Article
Full-text available
Alzheimer's disease (AD) is a neurodegenerative disease that involves multiple systems in the body. Numerous recent studies have revealed bidirectional crosstalk between the brain and bone, but the interaction between bone and brain in AD remains unclear. In this review, we summarize human studies of the association between bone and brain and provide an overview of their interactions and the underlying mechanisms in AD. We review the effects of AD on bone from the aspects of AD pathogenic proteins, AD risk genes, neurohormones, neuropeptides, neurotransmitters, brain‐derived extracellular vesicles (EVs), and the autonomic nervous system. Correspondingly, we elucidate the underlying mechanisms of the involvement of bone in the pathogenesis of AD, including bone‐derived hormones, bone marrow‐derived cells, bone‐derived EVs, and inflammation. On the basis of the crosstalk between bone and the brain, we propose potential strategies for the management of AD with the hope of offering novel perspectives on its prevention and treatment. Highlights The pathogenesis of AD, along with its consequent changes in the brain, may involve disturbing bone homeostasis. Degenerative bone disorders may influence the progression of AD through a series of pathophysiological mechanisms. Therefore, relevant bone intervention strategies may be beneficial for the comprehensive management of AD.
... ALP is a membrane-bound enzyme that stimulates osteoblast activity and bone mineralization. Osteoblasts secrete OCN and OPN that are also regarded as biomarkers of bone mineralization [30,31]. RUNX2 is an essential transcription factor for osteoblast differentiation [32]. ...
... The expression of collagen type 1 (Col I) and the osteoblast-specific extracellular matrix proteins, osteopontin (OPN), are reliable indicators of osteogenic differentiation 40,41 . Also, OPN is the most important bone biomarker responsible for the initiation and maintenance of bone mineralization 42,43 . We found that cells cultured on 3D-DPFS expressed significantly higher Col I and OPN compared to those on other scaffolds. ...
Article
Full-text available
Electrospinning is an effective method to fabricate fibrous scaffolds that mimic the ECM of bone tissue on a nano- to macro-scale. However, a limitation of electrospun fibrous scaffolds for bone tissue engineering is the structure formed by densely compacted fibers, which significantly impedes cell infiltration and tissue ingrowth. To address this problem, several researchers have developed numerous techniques for fabricating 3D fibrous scaffolds with customized topography and pore size. Despite the success in developing various 3D electrospun scaffolds based on fiber repulsion, the lack of contact points between fibers in those scaffolds has been shown to hinder cell attachment, migration, proliferation, and differentiation due to excessive movement of the fibers. In this article, we introduce a Dianthus caryophyllus-inspired scaffold fabricated using SIAC-PE, a modified collector under specific viscosity conditions of PCL/LA solution. The developed scaffold mimicking the structural similarities of the nature-inspired design presented enhanced cell proliferation, infiltration, and increased expression of bone-related factors by reducing fiber movements, presenting high space interconnection, high porosity, and controlled fiber topography.
... Increased ALPL activity indicates early regulation in osteogenesis, while the expression of SPP1 indicates its function in bone remodeling and periodontal regeneration. The RUNX2, a marker for osteoblast development and chondrocyte maturation, is overexpressed for effective osteogenic differentiation of PDLSCs [52][53][54]. This study takes a closer look at the lncRNA MALAT1 with in-vitro overexpression and knockdown plasmids. ...
Article
Full-text available
Objectives This study aimed to investigate the regulatory roles of lncRNA MALAT1, miR-124-3p, and IGF2BP1 in osteogenic differentiation of periodontal ligament stem cells (PDLSCs). Materials and methods We characterized PDLSCs by employing quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analyses to evaluate the expression of key osteogenic markers including ALPL, SPP1, and RUNX2. Manipulation of lncRNA MALAT1 and miR-124-3p expression levels was achieved through transfection techniques. In addition, early osteogenic differentiation was assessed via Alkaline phosphatase (ALP) staining, and mineral deposition was quantified using Alizarin Red S (ARS) staining. Cellular localization of lncRNA MALAT1 was determined through Fluorescence In Situ Hybridization (FISH). To elucidate the intricate regulatory network, we conducted dual-luciferase reporter assays to decipher the binding interactions between lncRNA MALAT1 and miR-124-3P as well as between miR-124-3P and IGF2BP1. Results Overexpression of lncRNA MALAT1 robustly promoted osteogenesis in PDLSCs, while its knockdown significantly inhibited the process. We confirmed the direct interaction between miR-124-3p and lncRNA MALAT1, underscoring its role in impeding osteogenic differentiation. Notably, IGF2BP1 was identified as a direct binding partner of lncRNA MALAT1, highlighting its pivotal role within this intricate network. Moreover, we determined the optimal IGF2BP1 concentration (50 ng/ml) as a potent enhancer of osteogenesis, effectively countering the inhibition induced by si-MALAT1. Furthermore, in vivo experiments utilizing rat calvarial defects provided compelling evidence, solidifying lncRNA MALAT1’s crucial role in bone formation. Conclusions Our study reveals the regulatory network involving lncRNA MALAT1, miR-124-3p, and IGF2BP1 in PDLSCs’ osteogenic differentiation. Clinical relevance These findings enhance our understanding of lncRNA-mediated osteogenesis, offering potential therapeutic implications for periodontal tissue regeneration and the treatment of bone defects.
Article
Objectives The value of biochemical markers of bone turnover (BTMs) in predicting survival and disease remains unclear. In a prospective study we evaluated the novel biomarkers for bone turnover sclerostin, dickkopf-1 (DKK-1), osteopontin (OPN), osteoprotegerin (OPG) and osteocalcin (OC), as well as a traditional biomarker, alkaline phosphatase (ALP) in relation to risk of mortality, cardiovascular events and fractures. Participants and Methods:Routine blood tests and serum BTMs, including ALP, were analyzed in patients with hip fracture n = 97, stroke n = 71 and healthy volunteers n = 83 (mean age 86, 83 and 77, respectively), followed for 7 years. Hazard Ratios (HR) were calculated for mortality, cardiovascular events and fractures in relation to these biomarkers. After adding the albumin-to-ALP ratio (AAPR) a post hoc analysis was performed. Results 120 participants died during the study. In the entire group of patients and volunteers (n = 251) higher AAPR (HR 0.28, 95 % CI 0.14–0.59, p < 0.001) was associated with decreased mortality. OPN and OPG were associated with mortality risk only in the univariate statistical analysis. HR for high AAPR in relation to new cardiovascular events was borderline significant (HR 0.29, 95 % CI 0.08–1.06, p = 0.061). None of the examined biomarkers were associated with new fractures, nor with an increased risk of a new cardiovascular event. Conclusions AAPR may be a better predictor of mortality than the more novel BTMs, and higher AAPR could be associated with longer life expectancy. Further studies should determine the clinical usefulness of AAPR as a biomarker of mortality and cardiovascular disease.
Article
The objectives of this study were to evaluate the contribution of additive and dominance genetic effects to the phenotypic variation of carcass quality traits and to identify the underlying genetic variants associated with these traits. A total of 3,958 Canadian crossbred beef cattle with phenotype and genotype data were used in two models: 1) additive and 2) joint additive and dominance genomic model which included fixed contemporary group, and covariates of slaughter age, and the eigenvectors of five principal components to account for population structure. Variance components and genome-wide association analyses were performed, and a 10% genome-wide false discovery rate (FDR) was applied to declare associations as significant. Genomic heritability ranged from 0.31±0.03 for ultrasound rib eye area to 0.46±0.05 for marbling score. Up to 10% dominance genetic variation was observed for ultrasound rib eye area and marbling score, indicating the contribution of dominance genetic effects to these trait variations. Eleven overlapping significant SNP associations were identified across the studied traits and models. The identified candidate genes (e.g. BTC, SPP1, and SEPSECS) have biological functions related to tissue growth and skeletal muscle development and can be further validated in other cattle populations to determine their usefulness for beef cattle genetic improvement.
Article
Full-text available
Over the last years non-alcoholic fatty liver disease (NAFLD) has grown into the most common chronic liver disease globally, affecting 17-38% of the general population and 50-75% of patients with obesity and/or type 2 diabetes mellitus (T2DM). NAFLD encompasses a spectrum of chronic liver diseases, ranging from simple steatosis (non-alcoholic fatty liver, NAFL) and non-alcoholic steatohepatitis (NASH; or metabolic dysfunction-associated steatohepatitis, MASH) to fibrosis and cirrhosis with liver failure or/and hepatocellular carcinoma. Due to its increasing prevalence and associated morbidity and mortality, the disease-related and broader socioeconomic burden of NAFLD is substantial. Of note, currently there is no globally approved pharmacotherapy for NAFLD. Similar to NAFLD, osteoporosis constitutes also a silent disease, until an osteoporotic fracture occurs, which poses a markedly significant disease and socioeconomic burden. Increasing emerging data have recently highlighted links between NAFLD and osteoporosis, linking the pathogenesis of NAFLD with the process of bone remodeling. However, clinical studies are still limited demonstrating this associative relationship, while more evidence is needed towards discovering potential causative links. Since these two chronic diseases frequently co-exist, there are data suggesting that anti-osteoporosis treatments may affect NAFLD progression by impacting on its pathogenetic mechanisms. In the present review, we present on overview of the current understanding of the liver-bone cross talk and summarize the experimental and clinical evidence correlating NAFLD and osteoporosis, focusing on the possible effects of anti-osteoporotic drugs on NAFLD.
Article
Full-text available
Background Over the last decade, we have been able to understand the role of various genetic processes involved in the oral diseases. However, In past few years, much of stresses has been paid to understand the regulation of these genetic processes controlled by the epigenetic mechanisms. Epigenetic processes regulate the gene activity without altering underlying DNA sequences, through mechanisms including DNA methylation, histone modifications, and non-coding RNAs. These epigenetic processes are significantly associated with various oral health problems including periodontitis, oral cancer. Highlights Different strategies like Genome-wide DNA methylation studies, microRNA based profiling studies, next-generation RNA sequencing analyses have been proven to be helpful in establishing the relationship among these epigenetic processes and gene expression. A few studies also suggest an emerging association of various epigenetic biomarkers in oral cancer and periodontal diseases. Conclusion Epigenetic mechanisms are highly sensitive and reversible, thereby offers significant potential for their use to develop novel diagnostic tools, pharmacotherapies and personalized based treatment care. However, there is lack of integrated approaches combining epigenomics with proteomics and genomics comparing the usefulness of epigenetic biomarkers in oral health research.
Article
Full-text available
The present study aimed to investigate the expression and regulation of extracellular signal‑regulated kinase (ERK)1/2 and p38 mitogen‑activated protein kinase (MAPK) signaling pathways in periodontal tissue remodeling of orthodontic tooth movement. Sprague Dawley rats with orthodontic tooth movement were generated. After tension stress for 1, 3, 5, 7 and 14 days, the protein and mRNA expression levels of ERK1/2 and p38 in periodontal tissue were determined by western blotting and reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR), respectively. Primary human periodontal ligament cells (hPDLCs) were separated and characterized. Following exposure to centrifugal force for 1, 2, 6, 8 and 12 h, the protein expression levels of ERK1/2 and p38 MAPK, and the mRNA expression levels of ERK1/2, p38 and osteogenesis associated‑genes [including alkaline phosphatase (ALP), osteopontin (OPN), collagen I (Col I), osteocalcin (OCN) and bone sialoprotein (BSP)] were measured. The protein expression levels of ERK1/2 and p38 MAPK in periodontal tissue and hPDLCs treated with stress were similar to those in the control groups. However, compared with the control, the phosphorylation and mRNA expression levels of the genes encoding ERK1/2 and p38 MAPK in orthodontic periodontal tissue and forced hPDLCs were elevated. These increases reached a peak at 5 days for orthodontic periodontal tissue and at 6 h for forced hPDLCs. In forced hPDLCs, the mRNA expression levels of ALP, OPN, Col I, OCN and BSP were notably and continuously upregulated in a time‑dependent manner. In addition, hPDLCs were treated with the ERK1/2 inhibitor, PD098059, and the p38 MAPK inhibitor, SB203580, and the mRNA expression levels of the osteogenesis associated‑genes were then measured using RT‑qPCR. Following treatment with the ERK1/2 inhibitor and p38 MAPK inhibitor, the mRNA expression levels of ALP, OPN, Col I, OCN and BSP were significantly downregulated. In conclusion, ERK1/2 and p38 MAPK signaling pathways may be positively and closely associated with periodontal tissue remodeling of orthodontic tooth movement.
Article
Full-text available
Objective: Genes, involved in the modulation of inflammatory response and bone remodeling, play a role in the development of post-orthodontic external apical root resorption (EARR). The aim of our study was to analyze possible associations between seven single nucleotide polymorphisms (SNPs) in interleukin-17A (IL-17), osteopontin (SPP1), purinoreceptor P2X7 (P2RX7) and tumor necrosis factor receptor superfamily member 11B (TNFRSF11B) genes and EARR in children after orthodontic treatment. Subjects and methods: This case-control study comprised 99 orthodontically treated patients (69 controls and 30 subjects with EARR). Genotype determinations of rs2275913, rs11730582, rs9138, rs208294, rs1718119, rs3102735 and rs2073618 were based on polymerase chain reaction using 5'nuclease TaqMan(®) assays. Results: While no significant differences were observed in allele or genotype frequencies of all seven studied SNPs, specific haplotype of P2RX7 (rs208294 and rs1718119) modified the risk of EARR development (P<0.05). In addition, the length of treatment with a fixed orthodontic appliance positively correlated with the presence of EARR (P<0.05). Conclusions: Although the effect of individual SNPs studied on the EARR development was not confirmed in the Czech population, complex analysis suggested that variability in the P2RX7 gene and the length of orthodontic treatment may be important factors contributing to the etiopathogenesis of post-orthodontic EARR. This article is protected by copyright. All rights reserved.
Article
Full-text available
Objective: To evaluate the expression of receptor activator of nuclear factor-kappa ligand (RANKL), osteoprotegerin (OPG), osteopontin (OPN), interleukin 1β (IL-1β), and transforming growth factor ß1 (TGF-ß1) in the gingival crevicular fluid (GCF) of teeth subjected to orthodontic forces released by aligners. Materials and methods: A total of 10 healthy, adult patients were selected to participate in this split-mouth study. The treatment plan was designed to obtain only one movement with the first aligner: distalization of a second molar. GCF samples were obtained from pressure and tension sites of the test tooth and from the mesiobuccal and distobuccal sites of the control tooth. The GCF sample volumes were measured using a Periotron 8000. Levels of TGF-β, IL-1β, RANKL, OPG, and OPN were measured by enzyme-linked immunosorbent assay. Results: IL-1β showed a significant increase at the pressure sites after 1 week and 3 weeks with respect to baseline (P < .05) compared with control sites. The kinetics of TGF-1β and OPN were characterized by a significant increase at the tension sites of the test teeth (P < .05) after 3 weeks from the application of orthodontic force. The RANKL level was significantly increased at pressure and tension sites after 1 hour and after 1 week from the application of the orthodontic forces (P = .023 and P = .043, respectively). Conclusions: An increased concentration of bone modeling and remodeling mediators at the pressure sites (IL-1β, RANKL) and tension sites (TGF-1β, OPN) was observed. These scenarios are compatible with previous in vivo and in vitro studies investigating the biological effects of orthodontic tooth movement.
Article
Full-text available
Aging diminishes bone formation engendered by mechanical loads, but the mechanism for this impairment remains unclear. Because Wnt signaling is required for optimal loading-induced bone formation, we hypothesized that aging impairs the load-induced activation of Wnt signaling. We analyzed dynamic histomorphometry of 5-, 12- and 22-month C57Bl/6JN mice subjected to multiple days of tibial compression and corroborated an age-related decline in the periosteal loading response on day 5. Similarly, 1 day of loading increased periosteal and endocortical bone formation in young-adult (5-mo) mice, but old (22-mo) mice were unresponsive. These findings corroborated mRNA expression of genes related to bone formation and the Wnt pathway in tibiae after loading. Multiple bouts (3-5 days) of loading up-regulated bone formation-related genes, e.g., Osx and Col1a1, but older mice were significantly less responsive. Expression of Wnt negative regulators, Sost and Dkk1, was suppressed with a single day of loading in all mice, but suppression was sustained only in young-adult mice. Moreover, multiple days of loading repeatedly suppressed Sost and Dkk1 in young-adult, but not in old tibiae. The age-dependent response to loading was further assessed by osteocyte staining for Sclerostin and LacZ in tibia of TOPGAL mice. After 1 day of loading, fewer osteocytes were Sclerostin-positive and, corroboratively, more osteocytes were LacZ-positive (Wnt active) in both 5- and 12-mo mice. However, while these changes were sustained after multiple days of loading in 5-mo mice, they were not sustained in 12-mo mice. Lastly, Wnt1 and Wnt7b were the most load-responsive of the 19 Wnt ligands. However, 4 hours after a single bout of loading, while their expression was upregulated 3-10 fold in young-adult mice, it was not altered in old mice. In conclusion, the reduced bone formation response of aged mice to loading may be due to failure to sustain Wnt activity with repeated loading. This article is protected by copyright. All rights reserved.
Article
Osteopontin (OPN) is a secreted glycoprotein with mineral‐ and cell‐binding properties that can regulate cell activities through integrin receptors. Previously, we identified an intracellular form of osteopontin with a perimembranous distribution in migrating fetal fibroblasts (Zohar et al., J Cell Physiol 170:88–98, 1997). Since OPN and CD44 expression are increased in migrating cells, we analyzed the relationship of these proteins with immunofluorescence and confocal microscopy. A distinct co‐localization of perimembranous OPN and cell‐surface CD44 was observed in fetal fibroblasts, periodontal ligament cells, activated macrophages, and metastatic breast cancer cells. The co‐localization of OPN and CD44 was prominent at the leading edge of migrating fibroblasts, where OPN also co‐localized with the ezrin/radixin/moesin (ERM) protein ezrin, as well as in cell processes and at attachment sites of hyaluronan‐coated beads. The subcortical location of OPN in these cells was verified by cell‐surface biotinylation experiments in which biotinylated CD44 and non‐biotinylated OPN were isolated from complexes formed with hyaluronan‐coated beads and identified with immunoblotting. That perimembranous OPN represents secreted protein internalized by endocytosis or phagocytosis appeared to be unlikely since exogenous OPN that was added to cell cultures could not be detected inside the cells. A physical association with OPN, CD44, and ERM, but not with vinculin or α‐actin, was indicated by immunoadsorption and immunoblotting of cell proteins in complexes extracted from hyaluronan‐coated beads. The functional significance of OPN in this complex was demonstrated using OPN−/− and CD−/− mouse fibroblasts which displayed impaired migration and a reduced attachment to hyaluronan‐coated beads. These studies indicate that OPN exists as an integral component of a hyaluronan‐CD44‐ERM attachment complex that is involved in the migration of embryonic fibroblasts, activated macrophages, and metastatic cells. J. Cell. Physiol. 184:118–130, 2000. © 2000 Wiley‐Liss, Inc.
Article
The periodontal complex is essential for tooth attachment and function and includes the mineralized tissues, cementum and alveolar bone, separated by the unmineralized periodontal ligament (PDL). To gain insights into factors regulating cementum-PDL and bone-PDL borders and protecting against ectopic calcification within the PDL, we employed a proteomic approach to analyze PDL tissue from progressive ankylosis knock-out (Ank−/−) mice, featuring reduced PPi, rapid cementogenesis, and excessive acellular cementum. Using this approach, we identified the matrix protein osteopontin (Spp1/OPN) as an elevated factor of interest in Ank−/− mouse molar PDL. We studied the role of OPN in dental and periodontal development and function. During tooth development in wild-type (WT) mice, Spp1 mRNA was transiently expressed by cementoblasts and strongly by alveolar bone osteoblasts. Developmental analysis from 14 to 240 days postnatal (dpn) indicated normal histological structures in Spp1−/− comparable to WT control mice. Microcomputed tomography (micro-CT) analysis at 30 and 90 dpn revealed significantly increased volumes and tissue mineral densities of Spp1−/− mouse dentin and alveolar bone, while pulp and PDL volumes were decreased and tissue densities were increased. However, acellular cementum growth was unaltered in Spp1−/− mice. Quantitative PCR of periodontal-derived mRNA failed to identify potential local compensators influencing cementum in Spp1−/− vs. WT mice at 26 dpn. We genetically deleted Spp1 on the Ank−/− mouse background to determine whether increased Spp1/OPN was regulating periodontal tissues when the PDL space is challenged by hypercementosis in Ank−/− mice. Ank−/−; Spp1−/− double deficient mice did not exhibit greater hypercementosis than that in Ank−/− mice. Based on these data, we conclude that OPN has a non-redundant role regulating formation and mineralization of dentin and bone, influences tissue properties of PDL and pulp, but does not control acellular cementum apposition. These findings may inform therapies targeted at controlling soft tissue calcification.
Article
Introduction: Bone turnover associated with orthodontic tooth movement is evidenced by increased bone turnover markers in gingival crevicular fluid (GCF). Postmenopausal women have an increased concentration of serum bone turnover markers. The filtrate of this serum makes up GCF, but little is known of the bone turnover around teeth in this cohort. The objective of this investigation was to compare the GCF bone turnover markers in premenopausal vs postmenopausal women receiving orthodontic treatment at baseline and at orthodontic activation. Methods: Twenty-eight women were enrolled in the study and separated into 2 groups: premenopausal (16) and postmenopausal (12). Bone turnover was evaluated by GCF at baseline and 24 hours after orthodontic appliance activation. GCF concentrations of RANKL and OPN were measured using ELISA. Baseline and change in concentrations were compared between groups. Results: Baseline RANKL and OPN were significantly different between the premenopausal and postmenopausal groups (P <0.05). Both markers increased significantly from baseline to 24 hours after orthodontic appliance activation in both groups (P <0.05). However, the response to orthodontic activation was not significantly different between groups. Conclusions: Although postmenopausal women have a different bone turnover profile at baseline than do their premenopausal counterparts, there is no difference in their response to orthodontic activation. This confers a level of security associated with orthodontic activation. Future studies are warranted to construct biomarker curves throughout orthodontic therapy.
Article
Objective: To determine whether orthodontic treatment with removable aligners vs fixed orthodontic appliances is associated with a different frequency of orthodontically induced external apical root resorption (OIEARR) when genetic, radiographic, and clinical factors are accounted for. Materials and methods: Three hundred seventy-two orthodontic patients treated with removable aligners (Invisalign) or fixed appliances were genetically screened for interleukin 1B gene (IL1B) (rs1143634), interleukin 1 receptor antagonist gene (IL1RN) (rs419598), and osteopontin gene (SPP1) (rs9138/rs11730582). Twelve clinical variables, potentially associated with OIEARR, were also considered. Subjects were divided according to the presence of radiographically determined OIEARR (>2 mm). The association between OIEARR and appliance type, and radiographic, clinical and genetic factors, was assessed using backward stepwise conditional logistic regression. Odds ratios (ORs) and 95% confidence intervals (CIs) were reported. Results: Reliability of the methods was adequate. Clinical case complexity (American Board of Orthodontics [ABO] Discrepancy Index) (OR: 1.032; 95% CI: 1.005-1.061; P = .021) and extent of incisor apical displacement in the sagittal plane (OR: 1.478; 95% CI: 1.285-1.699; P = .001) were associated with an increased OIEARR risk. After adjusting for associations between clinical/radiographic/genetic factors, there were no statistically significant differences with respect to OIEARR or type of orthodontic appliance used, whether removable aligners or fixed appliances (OR: 1.662; 95% CI: 0.945-2.924; P = .078). Only subjects homozygous for the T allele of IL1RN (rs419598) were more prone to OIEARR during orthodontic treatment (OR: 3.121; CI: 1.93-5.03; P < .001). Conclusions: A similar OIEARR predisposition was identified using either removable aligners (Invisalign) or fixed appliances.