ArticlePDF Available

The role of osteopontin in inflammatory processes

Wiley
Journal of Cell Communication and Signaling
Authors:

Abstract

Osteopontin (OPN) is a matricellular protein that mediates diverse biological functions. OPN is involved in normal physiological processes and is implicated in the pathogenesis of a variety of disease states, including atherosclerosis, glomerulonephritis, cancer, and several chronic inflammatory diseases. Through interactions with several integrins, OPN mediates cell migration, adhesion, and survival in many cell types. OPN also functions as a Th1 cytokine, promotes cell-mediated immune responses, and plays a role in chronic inflammatory and autoimmune diseases. Besides its function in inflammation, OPN is also a regulator of biomineralization and a potent inhibitor of vascular calcification.
RESEARCH ARTICLE
The role of osteopontin in inflammatory processes
Susan Amanda Lund &Cecilia M. Giachelli &
Marta Scatena
Received: 15 May 2009 / Accepted: 9 September 2009 /Published online: 2 October 2009
#The Author(s) 2009. This article is published with open access at Springerlink.com
Abstract Osteopontin (OPN) is a matricellular protein that
mediates diverse biological functions. OPN is involved in
normal physiological processes and is implicated in the
pathogenesis of a variety of disease states, including
atherosclerosis, glomerulonephritis, cancer, and several
chronic inflammatory diseases. Through interactions with
several integrins, OPN mediates cell migration, adhesion,
and survival in many cell types. OPN also functions as a
Th1 cytokine, promotes cell-mediated immune responses,
and plays a role in chronic inflammatory and autoimmune
diseases. Besides its function in inflammation, OPN is also
a regulator of biomineralization and a potent inhibitor of
vascular calcification.
Keywords Inflammation .Matricellular protein .
Osteopontin
Abbreviations
DC Dendritic cell
IFN Interferon
MMP Matrix metalloproteinase
OPN Osteopontin
PBMCs Peripheral blood mononuclear cells
Introduction
OPN is a secreted phosphorylated glycoprotein that
mediates diverse biological functions. Originally isolated
from bone, OPN was later shown to have a wider
distribution (Brown et al. 1992). In adults, OPN expression
is normally limited to the bone, kidney, and epithelial
linings, and is secreted in bodily fluids including milk,
blood and urine (Chen et al. 1993). In contrast to its
restricted distribution in normal tissue, OPN is strikingly
upregulated at sites of inflammation and tissue remodeling
(Liaw et al. 1998;OBrien et al. 1994). OPN exists both as
a component of the extracellular matrix and as a soluble
cytokine. Physiologically OPN is thought to regulate
biomineralization in bone tissue, and to reduce growth
and aggregation of calcium crystals in epithelial tissues
(Wesson et al. 2003). OPN has also been implicated in a
variety of disease states, where it mediates diverse cellular
functions such as adhesion, migration, and survival of
several different cell types, including regulating and
propagating inflammatory responses of macrophages, T-
cells, and dendritic cells. The pleiotropic nature of OPN
may reflect the various isoforms, post-translational mod-
ifications, and diversity of cell types which OPN can
interact with. Clinically, OPN plasma levels are correlated
with chronic inflammatory diseases such as Crohns disease
(Agnholt et al. 2007), cancer (El-Tanani et al. 2006),
atherosclerosis, aortic abdominal aneurysms (Golledge et
al. 2007), and autoimmune diseases including lupus
(Kariuki et al. 2009), multiple sclerosis (Comabella et al.
2005), and rheumatoid arthritis (Sennels et al. 2008). In this
review we will focus on the role of OPN in inflammation
biology.
OPN structure
OPN was originally isolated from bone and was later
independently identified as secreted phosphoprotein I
S. A. Lund :C. M. Giachelli :M. Scatena (*)
Department of Bioengineering, University of Washington,
Box 358056, Seattle, WA 98195, USA
e-mail: mscatena@u.washington.edu
J. Cell Commun. Signal. (2009) 3:311322
DOI 10.1007/s12079-009-0068-0
(SppI) and early T-lymphocyte activation 1 (Eta-1) (Senger
et al. 1989; Patarca et al. 1989). OPN is a member of the
SIBLING (Small Integrin-Binding Ligand, N-linked Gly-
coprotein) family of proteins, which map to human
chromosome 4 (Fisher et al. 2001). OPN is synthesized as
an approximately 32 kDa protein, but due to extensive post-
translational modifications its apparent molecular mass
ranges from 45 to 75 kDa (Kazanecki et al. 2007). OPN
possesses a negative charge due to a preponderance of
acidic amino acids and serine phosphorylation. OPN also
contains calcium binding sites and two putative heparin
binding domains (Kon et al. 2008). OPN can interact
directly with extracellular matrix proteins including fibro-
nectin (Mukherjee et al. 1995) and collagen type I (Chen et
al. 1992; Martin et al. 2004).
Adhesion motifs
OPN has multiple functional adhesive motifs, which allows
interactions with many cell types including smooth muscle
cells, endothelial cells, and inflammatory cells, thus
mediating a broad range of biological functions. The OPN
protein is poorly conserved among species (63% human to
mouse, 30% human to chicken) however its functional
domains are conserved. The highly conserved motifs
include: abundance of acidic residues, the RGD integrin
binding domain, similar phosphorylation and glycosylation
motifs, and at least one site of controlled proteolysis
(Bellahcene et al. 2008). The adhesive RGD domain of
OPN mediates interactions via α
v
β
1,
α
v
β
3
,α
v
β
5,
α
v
β
6,
α
8
β
1
, and α
5
β
1
integrins (Liaw et al. 1995; Yokosaki et al.
2005; Denda et al. 1998; Hu et al. 1995). Immediately C-
terminal to the RDG motif is a cryptic SVVYGLR
(SLAYGLR in mice) sequence that becomes exposed upon
cleavage with thrombin and mediates interactions with
α
9
β
1,
α
4
β
1
, and α
4
β
7
integrins (Yokosaki et al. 1999; Ito et
al. 2009; Green et al. 2001). Finally, the ELVTDFTDLPAT
(human OPN) domain of OPN has also been described to
bind to α
4
β
1
(Bayless and Davis 2001).
In addition to interacting with integrins, OPN has also
been reported to interact with CD44, the hyaluronic acid
receptor (Weber et al. 1996). A number of different CD44
isoforms exist due to alternative splicing of the 10 variant
exons and while OPN can bind some CD44 splice variants,
notably v6 and v7, OPN does not bind to the standard
isoform, CD44H (Smith et al. 1999; Katagiri et al. 1999).
Further, interactions between OPN and CD44 appears to be
mediated via β
1
integrins in an RGD independent manner
(Katagiri et al. 1999). Other studies indicate interactions
between the C-terminal fragment of thrombin cleaved OPN
and a CD44 variant (Weber et al. 2002). However, the
precise domain of OPN that interacts with CD44 has not
been identified. In addition, growing evidence suggests that
OPN is a major regulator of CD44 surface expression,
especially in osteoclasts (Chellaiah et al. 2003; Marroquin
et al. 2004).
Post-translational modifications
OPN is subject to extensive post-translational modifica-
tions including serine and threonine phosphorylation.
Phosphorylation is cell specific with phosphorylation
levels varying depending on the tissue type. OPN in
milk is highly phosphorylated with human milk OPN
containing 36 phosphate sites (Christensen et al. 2005).
Phosphorylation tends to occur in clusters separated by
stretches of unmodified residues. Normal rat kidney cells
can secrete both phosphorylated and non-phosphorylated
forms, indicating regulated control of phosphorylation
(Singh et al. 1990). In some cases, OPN function is tightly
controlled by phosphorylation state. Calcification of
smooth muscle cells in vitro is inhibited by native OPN,
but desphosphorylated or recombinant bacterially pro-
duced OPN has no effect on calcification (Jono et al.
2000). Similarly, in vivo in OPN-null mice, phosphorylat-
ed OPN, but not non-phosphorylated, prevents ectopic
calcification in a subcutaneous model of bioprosthetic
valve mineralization (Ohri et al. 2005). In contrast, the
adhesive activity of OPN is not dependent on post-
translational modifications, since bacterially derived re-
combinant OPN has been shown to support cell adhesion
in a wide variety of cell types (Gao et al. 2004;Xuanetal.
1994). OPN is also subject to sulfation (Nagata et al.
1989), glycosylation (Sorensen et al. 1995)andtrans-
glutamination (Beninati et al. 1994). Interestingly, poly-
merization of OPN by transglutaminase 2 has been
reported to increase the adhesive activity of OPN via the
α
9
β
1
integrin, independent of the SVVYGLR adhesion
domain (Nishimichi et al. 2009). Precise regulation of
OPN post-translational modifications may represent a
mechanism to control OPN function.
Proteolytic processing
The bioactivity of OPN can be further regulated by
proteolytic processing. OPN is a substrate for thrombin
and the matrix metalloproteinases, MMP-3 (stromelysin-1),
MMP-7 (matrilysin), MMP-2 and MMP-9 (Agnihotri et al.
2001; Dean and Overall 2007; Takafuji et al. 2007). Human
OPN contains three cleavage sites for MMPs; Gly
166
-
Leu
167
, Ala
201
-Tyr
202
, and Asp
210
-Leu
211
. The thrombin
cleavage site generating a RGD and SVVYGLR
(SLAYGLR in mice) is conserved in human and mice.
Thrombin cleavage of OPN, Arg
168
-Ser
169
in humans and
Arg
153
-Ser
154
in mice, reveals a cryptic SVVYGLR binding
domain capable of fostering interactions with α
9
β
1
(Smith
312 S.A. Lund et al.
and Giachelli 1998; Smith et al. 1996) and α
4
β
1
(Bayless
and Davis 2001) integrins. Thus, the known adhesive
functional domains of OPN are located in the thrombin
cleaved N-terminal fragment. Little is known about the role
of C-terminal fragments; however, as described above, it
has been suggested to contain a CD44 binding domain
(Weber et al. 2002; Takafuji et al. 2007).
Proteolytic processing may represent a way to locally
regulate the function of OPN as the functional properties of
cleaved OPN differ from those of the intact molecule. Of
particular interest, rather than mediating degradation and
inactivating OPN-mediated functions, proteolytic process-
ing of OPN can increase the biological activity of the
molecule (ORegan et al. 1999). OPN and MMPs are co-
localized during wound healing and tumorigenesis, indicat-
ing there may be an in vivo role for proteolyzed forms of
OPN (Senger et al. 1994). Few studies suggest that OPN
fragments may play a functional role in vivo. Enhanced
production of the thrombin cleaved form of OPN is found
in the synovial fluid of patients with rheumatoid arthritis
(Ohshima et al. 2002) and the cryptic SLAYGLR motif was
found to play an essential role in a murine and a primate
model of rheumatoid arthritis (Yamamoto et al. 2003,
2007). An antibody against the SLAYGLR sequence
inhibited inflammatory cell influx into arthritic joints and
attenuated the severity of disease. The human OPN derived
peptide SVVYGLR has also been found to induce
angiogenesis in vitro and in vivo (Hamada et al. 2003,
2007). Several in vitro studies have demonstrated that the
N-terminal fragments generated both by thrombin cleavage
and MMP cleavage induced enhanced adhesion when
compared to the full length molecule. This appears to be
due mostly to increased activity of the RGD site, perhaps
an indication of conformational change resulting in higher
affinity binding (Senger and Perruzzi 1996; Smith and
Giachelli 1998; Agnihotri et al. 2001). The SVVYGLR
cryptic domain exposed following thrombin cleavage is
also able to induce adhesion and migration through the α
4
and α
9
integrins (Smith and Giachelli 1998). However, the
α
9
-dependent adhesion and migratory functions are com-
pletely lost in the N-terminal MMP generated fragment, as
Arg
168
seems to be required for α
9
-dependent binding
(Yokosaki et al. 2005; Ito et al. 2009). In contrast, α
4
-
dependent adhesion and migratory functions are only
partially lost in the N-terminal MMP generated fragment
(Ito et al. 2009). The C-terminal fragment of OPN
generated by thrombin and MMP cleavage does not contain
any integrin adhesive domains, it does not mediate
adhesion when presented in immobilized form to cells
and, in contrast, it appears to suppress OPN mediated
adhesion and migration in monocyte-derived cells (Gao et
al. 2004; Smith et al. 1996; Maeda et al. 2001; Takahashi et
al. 1998)
Intracellular OPN
An intracellular form of OPN (iOPN) has been reported to
be expressed in dendritic cells and macrophages (Shinohara
et al. 2006,2008a,b; Zohar et al. 2000). Studies by
Shinohara et al. suggest that the intracellular form of OPN
is generated due to translation initiation downstream of the
usual start site in bone marrow-derived DCs and transfected
293T cells. Utilization of this downstream start site
generates the truncated iOPN form that lacks the N-
terminal signal sequence and consequently localizes to the
cytoplasm, where it may associate with TLR9 and the
MyD88 adaptor molecule (Shinohara et al. 2008a).
The interaction of iOPN with MyD88 appears to activate
the transcription factor IRF7 and to induce expression of
IFN-αultimately leading to Th1 cell-mediated immunity
and pro-inflammatory responses (Shinohara et al. 2006).
The same group has also suggested that iOPN expression in
conventional DCs is permissive for Th17 T cell responses
(Shinohara et al. 2008b). Th17 T cells are a subset of T
helper cells producing IL-17. They are considered devel-
opmentally distinct from Th1 and Th2 cells and are thought
to play a key role in autoimmune diseases including the
tissue injury associated with these conditions (Steinman
2007). Thus iOPN expression may allow for autoimmune
type disease progression. Indeed, OPN accelerates the
progression of experimental autoimmune encephalomyelitis
(EAE), a model of multiple sclerosis (Chabas et al. 2001;
Hur et al. 2007; Jansson et al. 2002; Shinohara et al. 2006,
2008b). Others have found that iOPN may associate with
the intracellular domain of CD44 and with the ezrin/
radixin/moesin (ERM) protein ezrin. iOPN in this context
may modulate cytoskeletal rearrangements important for
macrophage migration and osteoclast fusion (Zhu et al.
2004; Zohar et al. 2000).
A summary of the structural features of OPN is shown in
Fig. 1.
The role of OPN in inflammation
OPN regulates the immune system at many different levels. It
serves as a chemotactic molecule to promote the migration of
inflammatory cells to the wound site and acts as an adhesive
protein to retain cells at the site. OPN also functions as a pro-
inflammatory cytokine and can modulate the immune
response by enhancing expression of Th1 cytokines and
matrix degrading enzymes (Weber et al. 2002; Bruemmer et
al. 2003). OPN plays a pivotal role in T cell and macrophage
responses during cell mediated immune responses against
bacterial and viral pathogens (Ashkar et al. 2000). More
recently, OPN has also been shown to modulate dendritic
cell responses and neutrophil chemotaxis.
The role of osteopontin in inflammatory processes 313
Macrophages
OPN is not expressed in circulating monocytes, but is
dramatically upregulated during macrophage differentiation
and constitutes one of the major macrophage products
(Krause et al. 1996). OPN is known to be induced in
macrophages by several inflammatory cytokines, including
TNF-α,IL-1β,IFN-γ, and IL-6, and other factors
including angiotensin-II, oxidizedLDL, and phorbol-ester
are known inducers of OPN in macrophages (Nakamachi et
al. 2007; Ogawa et al. 2005; Bruemmer et al. 2003). More
recently, Liver X Receptor and Peroxisome proliferator-
activated receptor αantagonists have been shown to
suppress OPN expression (Nakamachi et al. 2007; Ogawa
et al. 2005). Functionally OPN plays a key role in
macrophage biology by regulating migration, survival,
phagocytosis, and pro-inflammatory cytokine production
(Bruemmer et al. 2003; Nyström et al. 2007).
We and others have shown that OPN serves as a potent
chemoattractant for macrophages (Bruemmer et al. 2003;
Persy et al. 2003; Giachelli et al. 1998; Panzer et al. 2001).
Functional inhibition of OPN and genetic ablation of OPN
in mice greatly impair macrophage recruitment in several
models of acute inflammation. In OPN-null mice acute
macrophage infiltration was greatly diminished com-
pared to wild-type mice in an obstructed kidneys model
(Ophascharoensuk et al. 1999), and in a thioglycollate
induced peritonitis model (Bruemmer et al. 2003). Further,
purified OPN induced macrophage accumulation when
injected in rat dermis and following intradermal injection
of N-formyl-met-leu-phe (FMLP), a potent macrophage
chemotactic peptide. The OPN effect was neutralized by
an anti-OPN blocking antibody (Giachelli et al. 1998). In a
rat model of heart injury OPN was highly expressed in the
granulation tissue associated macrophages, and it was
downregulated with healing progression and formation of
the fibrotic scar (Murry et al. 1994). Wound healing studies
in mice also indicate that OPN is expressed during the
acute inflammatory phase at very high levels in infiltrating
leukocytes and other cell types where it appears to regulate
leukocyte infiltration and activation as well as proper matrix
organization (Liaw et al. 1998). Interestingly, downregulation
of OPN at the wound site with antisense mRNA diminished
macrophage infiltration and accelerated wound healing
(Mori et al. 2008).
OPN also modifies chronic inflammatory responses.
Chronic inflammation is characterized by the persistence
of macrophages at sites of injury and disease. Deficits in
macrophage accumulation have been noted in OPN-null
mice when challenged with chronic inflammatory condi-
tions, including atherosclerosis, delayed-type hypersensi-
tivity (Yu et al. 1998; Ashkar et al. 2000), granulomatous
disease (Weber et al. 2002; Nau et al. 1999), and
biomaterial implantation (Steitz et al. 2002; Tsai et al.
2005). These data suggest that OPN may be particularly
important in promoting migration and retention of macro-
phages at sites of acute and chronic inflammation. We have
also shown that OPN regulates foreign body giant cell
(FBGC) formation in vitro and in vivo. In a recent paper we
described that despite the defect in macrophage recruitment,
OPN-null mice formed more FBGCs on the surface of the
implant. In vitro, OPN inhibited macrophage fusion to form
FBGCs in a dose dependent manner (Tsai et al. 2005).
In vitro, OPN-null macrophages exhibit reduced basal
migration and impaired migration towards MCP-1, despite
the fact that wild type and OPN-null macrophages express
comparable levels of CCR-2, the MCP-1 receptor. This may
be a consequence of a lack of a permissive pro-migratory
substrate, and the reduced expression of CD44 observed in
OPN-null macrophages. CD44 is well known to be
essential for macrophage migration (Marcondes et al.
2008), and its expression is upregulated by OPN in
macrophages (Chellaiah et al. 2003). Macrophages from
OPN-null mice are also more susceptible to programmed
cell death (Bruemmer et al. 2003). Together with impaired
migration, macrophage apoptosis may further contribute the
N
H2COOH
1169 300
Ca2+ binding
domain
RGD
(αvβ3, αvβ5,
αvβ1, α8β1)
Thrombin cleavage site
SVVYGLR
(α9β1, α4β1, α4β7)
MMP cleavage site
(CD44)
P P P P P P P P P PP
Ca2+ binding
domain
Fig. 1 OPN structural features.
The cell adhesive domains are
indicated in color. The known
specific integrin receptors for
each adhesion domain are also
shown in color. The calcium
(Ca
2+
) binding domains are
shown in yellow and other ma-
trix binding domains are in
black. Also shown are known
phosphorylation sites (P).
Arrows indicate known cleavage
sites for thrombin and MMPs.
Thrombin cleavage site in blue
and MMPs cleavage site in
green
314 S.A. Lund et al.
impaired macrophage accumulation observed in OPN-null
mice in response to acute and chronic inflammatory stimuli.
In addition to regulating macrophage migration, OPN
can also modulate the cytokine production by macro-
phages. OPN stimulates production of IL-12 while
inhibiting the production of IL-10, thereby promoting
Th1 cell mediated responses (Weber et al. 2002;Ashkar
et al. 2000). Interestingly, these results were mediated by
different receptors. IL-12 production was mediated via an
N-terminal fragment interaction with α
v
β
3
integrin, while
IL-10 was inhibited via a C-terminal fragment, possibly
via the CD44 receptor. OPN regulation of IL-12 and IL-10
were also demonstrated in vivo in an angiotensin II
(AngII)-accelerated model of atherosclerosis. In this
model, Bruemmer at al. showed reduced expression of
IL-12 and an increased expression of IL-10 in ApoE-/-
OPN-/- AngII treated mice compared to ApoE-/-OPN+/+
AngII treated control mice by RT-PCR of whole mouse
aortas. These results correlated with less macrophage rich
lesions and lower expression of the macrophage marker
CD68 (Bruemmer et al. 2003).
While OPN is generally classified as a pro-inflammatory
cytokine, it also appears to have anti-inflammatory effects.
OPN is a potent trans-repressor of inducible nitric oxide
synthase (iNOS) expression in macrophages (Rollo et al.
1996). OPN represses inducible nitric oxide synthase
(iNOS) by increasing Stat1 ubiquitination and proteasome
mediated degradation of Stat1, consequently inhibiting
Stat1 mediated iNOS transcription and protein expression
(Gao et al. 2007). NO feedback inhibits its own synthesis
by increasing transcription of OPN (Guo et al. 2001). OPN
inhibition of NO may be particularly relevant for tumor cell
evasion of inflammation (Wai et al. 2006; Crawford et al.
1998).
Finally, a recent study suggests that OPN may play a role in
macrophage differentiation. Using siRNA to stably silence the
expression of OPN in RAW 264.7 cells, Nystrom et al.
showed that OPN silenced cells displayed an altered pheno-
type with monocyte-like characteristics (Nystrom et al. 2007).
Further, OPN silenced cells had decreased expression of
macrophage scavenger receptor A type 1 (Msr-1), a
macrophage differentiation marker. While these studies are
intriguing, the phenotype could not be rescued by the
addition of exogenous OPN suggesting non-receptor medi-
ated effects or iOPN may be involved.
Together, these data suggest that OPN may be
particularly important in promoting migration and
retention of macrophages at sites of acute and chronic
inflammation by regulating multiple macrophage func-
tions. These studies also emphasize the importance of
macrophage-derived OPN in the regulation of OPNs
functions, suggesting that macrophages are both a
source and target of OPN.
Neutrophils
While much is known about the role of OPM
in macrophage biology, relatively few studies have ex-
plored the function of OPN in neutrophils. Neutrophils
express low levels of OPN (Koh et al. 2007). However,
OPN is important for the recruitment and migration of
neutrophils, as neutrophils from OPN-null mice display
reduced chemotaxis toward fMLP and in vivo the recruit-
ment of neutrophils to the peritoneal cavity in response to
sodium periodate is impaired in OPN-null mice (Koh et al.
2007). OPN-null mice also have impaired neutrophil
infiltration into liver when challenged with concanavalin
A induced hepatitis (Diao et al. 2004). Despite these defects
in migration and chemotaxis, OPN-null neutrophils do not
display reduced destructive capacity in terms of phagocy-
tosis, the generation of reactive oxygen species, or cytokine
production (Koh et al. 2007). Recent reports indicate that
polymeric OPN interacts with α
9
β
1
on neutrophils and
serves as a potent neutrophil chemoattractant (Nishimichi et
al. 2009).
T-cells
OPN is also known as Eta-1 (early T lymphocyte activation
gene 1) for its high expression in activated T cells and it
plays an important role in the induction of cell mediated
immune responses through the regulation of T cells.
Following activation, naïve CD4 T cells can differentiate
towards Th1, Th2, or Th17 cells which differ in effector
function. The development of Th1 cells leads to cell-
mediated immunity while development of Th2 cells
provides humoral immunity. Th17 cells are associated with
autoimmunity. OPN is not expressed in naïve T-cells but it
is strongly upregulated in response to T cell receptor
ligation (Shinohara et al. 2005). OPN functions in T cells
by mediating migration, adhesion, and co-stimulating T cell
proliferation (ORegan et al. 1999; Patarca et al. 1993).
Shinohara et al. have shown that OPN gene expression in T
cells is controlled by T-bet, a transcription factor that
promotes CD4+ T helper cell lineage commitment to Th1
(Shinohara et al. 2005). Further, T-bet-dependent expres-
sion of OPN in T cells is essential for efficient skewing of
CD4 T and CD8 T cells toward the Th1 and type 1 CD8 T
cell (Tc1) pathway, respectively (Shinohara et al. 2005). In
vivo, OPN-null mice display impaired Th1 responses to the
intracellular bacterium Listeria monocytogenes and the viral
pathogen HSV1 (herpes simplex virus type 1) both of
which depend on the induction of IL-12 for protection
(Ashkar et al. 2000). Indeed, mice deficient in OPN have
decreased IL-12 and IFN-γproduction, while IL-10 levels
are enhanced (Bruemmer et al. 2003). Further studies have
shown that OPN regulates CD3-mediated T cell expression
The role of osteopontin in inflammatory processes 315
of IFN-γand CD40L (ORegan et al. 2000), which in turn
stimulates IL-12 production from leukocytes. Together
these findings suggest that OPN may play a role in
polarizing early Th1 responses. More recently, OPN has
also been shown to regulate IFN-γand IL-17 production by
T cells in an α
v
β
3
-dependent manner and to dampen IL-10
in a CD44-dependent manner (Murugaiyan et al. 2008).
This appears to be important for the progression of
experimental autoimmune encephalomyelitis (EAE), a
murine model of multiple sclerosis. The same authors
found that during EAE, OPN expression was elevated in
DCs both in the periphery and in the CNS (Murugaiyan et
al. 2008). This correlated with the increased expression of
OPN in DCs and increased expression of OPN receptors
CD44 and α
v
β
3
on T cells isolated from patients affected
by multiple sclerosis (Murugaiyan et al. 2008). These
results may indicate that OPN produced by DCs in EAE is
linked to the production of IL-17 (Murugaiyan et al. 2008;
Shinohara et al. 2008b). Recent studies indicate that OPN
enhances the survival of autoreactive T cells in a NF-κB-
dependent manner. Enhanced OPN-induced T cell survival
also appears to promote progression of EAE (Hur et al.
2007). In addition, an anti-OPN antibody has been shown
to promote apoptosis of activated T cells, particularly CD4+
T cells, by inhibiting activation of NF-κB and by altering
the balance between the proapoptotic proteins, Bim and
Bax, and the antiapoptotic protein, Bcl-2, in a model of
rheumatoid arthritis (Fan et al. 2008).
Finally, a subset of lymphocytes bridging innate and
adaptive immunity called invariant natural killer T (iNKT)
cells has also been shown to be regulated by OPN. Diao et
al. found that the number of peripheral iNKT cells was
significantly reduced in OPN-deficient mice compared with
wild-type mice. This appeared to be consequence of
impairment of intrathymic iNKT cell maturation and
significant alteration of iNKT cell function as well (Diao
et al. 2004).
Dendritic cells
OPN plays a key role in DC maturation, migration, and
polarization. Interestingly, immature IL-4 and GM-CSF
differentiated DCs express high levels of OPN, and OPN
production decreases when DCs are stimulated with LPS
and CD40L (Kawamura et al. 2005; Schulz et al. 2008).
These findings suggest differential regulation of OPN in
DCs and macrophages. During macrophage differentiation,
OPN expression increases as monocytes adhere and
differentiate into macrophages (Krause et al. 1996).
In vitro OPN stimulates DC migration in a dose
dependent manner in both the presence and absence of
divalent cations, and studies with blocking antibodies have
indicated a role for the OPN receptors α
v
β
3
and CD44 in
OPN induced DC migration (Weiss et al. 2001). In vivo,
DCs in OPN-null mice display a defect in DC trafficking to
the lymph nodes resulting in reduced contact hypersensi-
tivity responses (Weiss et al. 2001). Despite the influence of
OPN on DC migratory capacity, treatment with recombi-
nant OPN does not affect the expression of CCR5 and
CCR7, chemokine receptors which are involved in DC
migration (Schulz et al. 2008). Recently, it has also been
suggested that OPN modulates different subset of DCs in
the airway hypersensitivity reaction (a model of asthma). It
appears that OPN increased the reaction during primary
sensitization but decreased the reaction in the challenge
phase, perhaps by mediating differential recruitment of
different DCs (plamacytoid and conventional) subsets to the
lymph nodes (Xanthou et al. 2007).
OPN also influences DC cytokine production. In co-
culture systems, OPN induces DC secretion of TNF-αand
IL-12p70 which stimulates secretion of IFN-γby T-cells
(Renkl et al. 2005). By augmenting DC production of IL-
12, OPN can enhance Th1 polarization.
As mentioned earlier, recent studies illustrate the critical
role of intracellular OPN in IFN-αproduction by plasma-
cytoid DCs, a specialized subset of DCs which produce
high levels of type I interferons upon stimulation
(Shinohara et al. 2006). IFN-αproduced by pDCs activates
NK cells, and consequently OPN deficient mice display
impaired IFN-αdependent natural killer cell responses.
Further, iOPN appears to decrease IL-27 in conventional
DCs, leading to increases in Th17 responses (Shinohara et
al. 2008b).
Other biological functions of OPN
Wound healing
OPN is a key cytokine regulating tissue repair. OPN is
present at sites of wound healing where it serves as a
chemotactic molecule to recruit inflammatory cells to the
site of injury. Wound healing studies in OPN-null mice
have elucidated the role of OPN in tissue repair. Compared
to wild type mice, incisional wounds made in OPN-null
mice displayed alterations in the matrix architecture
especially collagen fiber diameter, and had more residual
debris (Liaw et al. 1998). Furthermore, macrophages at the
wound site in OPN-null mice expressed higher levels of
mannose receptor, suggesting OPN may contribute to
macrophage polarization and thus regulate healing
responses. Mannose receptor expression in macrophages is
associated with reduced pro-inflammatory (IL-1, IL-6,
IL-12, and TNF-α) cytokine secretion, upregulation of
pro-healing molecules (IL-10 and TGF-β), and certain
phagocytic receptors (Sica et al. 2008).
316 S.A. Lund et al.
A more recent study by Mori et al. explored the use of
local OPN knockdown at the site of wound healing by
delivering antisense oligodeoxynucleotides from a drug
delivery polymer gel. Consistent with the finding in OPN-
null mice, in OPN knockdown wounds the diameter of
collagen fibrils was also smaller than in control wounds.
Further, OPN knockdown hindered the migration of
inflammatory cells to the wound site and resulted in
accelerated healing and a reduction in granulation tissue
formation and scarring (Mori et al. 2008). Whether the
infiltrated macrophages in OPN knockdown wounds also
had a less pro-inflammatory and more pro-healing pheno-
type was not established, however, this may be behind the
observed accelerated healing. The reduced size of the
collagen fibers that was observed in the OPN-null and
OPN knockdown wounds may be related to the finding that
OPN has been shown to bind directly to collagen type I
(Chen et al. 1992), and to interact with collagen types II,
III, IV, and V (Bulter et al. 1995). Further, it has recently
been shown that OPN is necessary for TGF-β1-induced
myofibroblast differentiation and OPN-null fibroblasts
exhibited less spreading, less resistance to detachment,
and a reduction in collagen gel contraction (Lenga et al.
2008). These studies indicate a role for OPN in promoting
proper collagen organization and regulating ECM and
myofibroblast interactions.
Vascular disease
To date, several studies have investigated the role of OPN in
the progression of atherosclerosis. In hyperlipidemic apoE-
deficient mice, Matsui et al, showed that osteopontin
deficiency significantly reduces atherosclerotic lesion size in
female ApoE-/-OPN-/- mice compared to ApoE-/-OPN+/+
mice after 36 weeks on a normal chow diet (Matsui et al.
2003). Similarly, studies in ApoE/LDLreceptor/OPN triple
knockout mice showed that OPN deficiency resulted in
decreased atherosclerotic lesion size and an increase in the
number of apoptotic cells in lesions (Strom et al. 2004).
Bone marrow transplantation studies in an angiotensin II-
accelerated model of atherosclerosis indicated that leukocyte
derived OPN contributes to OPN mediated development of
atherosclerosis (Bruemmer et al. 2003). These studies
suggest that OPN promotes macrophage accumulation and
retention in the atherosclerotic lesions, thus contributing to
the chronicity of the disease.
OPN also modulates other vascular cells associated with
vascular disease. In human atherosclerotic lesions, OPN is
expressed in smooth muscle cells (SMC), angiogenic endo-
thelial cells, and macrophages and it is re-expressed in SMCs
associated with human restenotic lesions (Giachelli et al.
1993; Panda et al. 1997). Consistently, animal models have
confirmed the role of OPN in promoting SMC migration and
proliferation (Liaw et al. 1994; Isoda et al. 2002). All these
data indicate that during injury, OPN enhances the prolifer-
ation, migration, and accumulation of smooth muscle and
endothelial cells involved in repair and remodeling processes
of the vasculature.
Cancer
OPN is highly expressed in transformed cells and is found
in a variety of cancers (Wai and Kuo 2008). OPN
overexpression can confer metastatic phenotype to non-
metastatic, benign transformed cells, and increased OPN
expression correlates with tumor progression, poor progno-
sis and increased invasiveness. In metastatic models, OPN
has been shown to induce matrix proteases MMP2 and uPA
in an integrin-dependent manner (Mi et al. 2006). Further,
OPN has been shown to bind and activate MMP3. Thus, the
ability of OPN to stimulate migration and matrix break-
down could contribute to invasiveness and to the metastatic
potential of tumors cells. OPN may also promote tumori-
genesis and metastasis by inhibiting apoptosis of tumor
cells (Zhao et al. 2008), and by stimulating neovasculariza-
tion (Wai and Kuo 2004). Finally, OPN is widely expressed
by macrophages, which infiltrate tumor tissue (Brown et al.
1994; Chambers et al. 1996). Macrophage-derived OPN
functions as a chemoattractant and was associated with
reduced tumor burden while tumor-derived OPN appeared
to inhibit macrophage function and enhance tumor growth
(Crawford et al. 1998). It is possible that tumor cell-derived
OPN may enhance cancer cell survival by downregulating
iNOS expression and NO production in macrophages. OPN
is currently being studied as a potential biomarker for
cancer and there is interest in targeting OPN as a
therapeutic treatment for cancer.
Biomineralization
OPN is one of the most abundant non-collagenous proteins
in bone. Because of its abundance in bone, OPN has been
studied as a regulator of biomineralization. OPN is a potent
inhibitor of mineralization, prevents ectopic calcification,
and is an inducible inhibitor of vascular calcification (Steitz
et al. 2002). OPN binds hydroxyapatite and calcium ions
thereby physically inhibiting crystal formation and growth
in vivo.Studies in OPN-null mice have shown that OPN-/-
bones are hypermineralized, with increased mineral content
and crystal size (Boskey et al. 2002). OPN also plays a role
in osteoclast differentiation and osteoblast recruitment and
function (Rittling et al. 1998). OPN functions in osteoclast
migration to sites of resorption and is crucial for normal
resorption and bone turnover (Chellaiah et al. 2003).
OPN appears also to be an important regulator of vascular
calcification and is associated with mineralized deposits in
The role of osteopontin in inflammatory processes 317
humans (Giachelli et al. 1993). In mice, OPN levels are
greatly elevated in the spontaneously mineralizing arteries of
MGP
-/-
mice and we have recently shown that OPN is major
inducible inhibitor of arterial medial calcification in this
system (Steitz et al. 2002). Vascular calcification is now
recognized as a marker of atherosclerotic plaque burden as
well as a major contributor to loss of arterial compliance and
increased pulse pressure seen with age, diabetes, and renal
insufficiency. These findings suggest that OPN may be an
important inhibitor of arterial mineral deposition under
conditions of injury and disease, and that strategies to
replenish OPN might be useful to prevent or treat ectopic
calcification, including vascular calcification.
Conclusions
OPN is emerging as a key regulator of immune cell biology.
Most of the evidence indicates that OPN is transiently
expressed in leukocytes during acute inflammation. How-
ever, persistence of OPN expression by immune cells
exacerbates chronic inflammatory diseases. Clinically, this
is manifested by increased OPN plasma levels in Crohns
disease (Agnholt et al. 2007), cancer (El-Tanani et al.
2006), atherosclerosis, and autoimmune diseases including
lupus (Kariuki et al. 2009), multiple sclerosis (Comabella et
al. 2005), and rheumatoid arthritis (Sennels et al. 2008).
Mechanistically, OPN appears to regulate innate immune
cells (macrophages and DCs) and adaptive immune cells
(T cells) at multiple levels (see Fig. 2). Recent data point to
a role for OPN in the regulation of cross-talk between DCs
and T cells and their subsequent polarization in Th1 and
more recently in Th17 cells.
In vitro and in vivo studies show that both the thrombin
and MMP proteolytically cleaved OPN fragments possess
higher activity than the full-length form. In addition, at least
the thrombin cleaved fragment also gains a new cell
interacting domain (SVVYGLR). Antibodies specifically
reacting toward the SVVYGLR (human and primate) or the
SLAYGLR (murine) sequences have been shown effective
in ameliorating rheumatoid arthritis symptoms in non-
human primates and mice (Yamamoto et al. 2003,2007).
INFγ Th1 polarization
OPN
DCs
T-Cells
macrophages
migration
activation
survival
Accumulation
IL-12
IL-10
Th1 polarization
migration
activation
IL-12 TNFα
Th1 polarization
survival
IL-17
Th17 polarization
EAE autoimmunity
iOPN
INFα
Th1 polarization
OPN fragments
N-terminal OPN
SVVYGLR
Rheumatoid
Arthritis
Angiogenesis
C-terminal OPN
?
migration
proliferation
Fig. 2 OPN regulation of immune and inflammatory cells. OPN is
secreted and modulates the function of macrophages, DCs and T
Cells. OPN may induce macrophage accumulation by promoting
migration and survival. Further, OPN induces IL-12 and inhibits IL-10
in macrophages, thus, propagating a Th1 response. In DCs OPN
appears to modulate their function as an extracellular soluble cytokine
and also as an intracellular molecule (iOPN). Both OPN forms appear
to induce Th1 polarization. Extracellular OPN appears to induce
expression of IL-12 and TNF-α, and iOPN appears to regulate the
production of INF-α. In T cells OPN induces migration, proliferation,
survival, and IL-17 secretion. These two latter functions have been
correlated with Th17 responses and autoimmunity. Further, OPN
appears to induce IFN-γsecretion by T cells thus propagating a Th1
response. Finally, the N-terminal OPN fragment (containing the
activated adhesive domain SVVYGLR) may be important in the
propagation of rheumatoid arthritis
318 S.A. Lund et al.
Therefore, during inflammation, it is likely that the
secreted, less potent, full-length OPN is rapidly cleaved
and thus activated. Understanding differences in the
mechanisms and structure/function relationships governing
the proinflammatory properties of OPN could help create
specific therapeutics aimed at targeting chronic inflamma-
tory diseases selectively.
Acknowledgements Dr. Giachellis laboratory is funded by NIH
grants HL62329, HL081785, and HL18645, and grant #2361524 from
the Washington State Life Science Discovery Fund.
Open Access This article is distributed under the terms of the
Creative Commons Attribution Noncommercial License which per-
mits any noncommercial use, distribution, and reproduction in any
medium, provided the original author(s) and source are credited.
References
Agnholt J, Kelsen J, Schack L, Hvas CL, Dahlerup JF, Sorensen ES
(2007) Osteopontin, a protein with cytokine-like properties, is
associated with inflammation in Crohnsdisease.ScandJ
Immunol 65(5):453460
Agnihotri R, Crawford HC, Haro H, Matrisian LM, Havrda MC, Liaw L
(2001) Osteopontin, a novel substrate for matrix metalloproteinase-3
(stromelysin-1) and matrix metalloproteinase-7 (matrilysin). J Biol
Chem 276(30):2826128267
Ashkar S, Weber GF, Panoutsakopoulou V, Sanchirico ME, Jansson
M, Zawaideh S, Rittling SR, Denhardt DT, Glimcher MJ, Cantor
H (2000) Eta-1 (osteopontin): an early component of type-1 (cell-
mediated) immunity. Science 287(5454):860864
Bayless KJ, Davis GE (2001) Identification of dual alpha4 beta1
integrin binding sites within a 38 amino acid domain in the N-
terminal thrombin fragment of human osteopontin. J Biol Chem
276(16):1348313489
Bellahcene A, Castronovo V, Ogbureke KU, Fisher LW, Fedarko NS
(2008) Small integrin-binding ligand N-linked glycoproteins
(SIBLINGs): multifunctional proteins in cancer. Nat Rev Cancer
8(3):212226
Beninati S, Senger DR, Cordella-Miele E, Mukherjee AB,
Chackalaparampil I, Shanmugam V, Singh K, Mukherjee BB
(1994) Osteopontin: its transglutaminase-catalyzed posttrans-
lational modifications and cross-linking to fibronectin. J
Biochem 115(4):675682
Boskey AL, Spevak L, Paschalis E, Doty SB, McKee MD
(2002) Osteopontin deficiency increases mineral content and
mineral crystallinity in mouse bone. Calcif Tissue Int 71
(2):145154
Brown LF, Berse B, Van de Water L, Papadopoulos-Sergiou A,
Perruzzi CA, Manseau EJ, Dvorak HF, Senger DR (1992)
Expression and distribution of osteopontin in human tissues:
widespread association with luminal epithelial surfaces. Mol Biol
Cell 3(10):11691180
Brown LF, Papadopoulos-Sergiou A, Berse B, Manseau EJ, Tognazzi
K, Perruzzi CA, Dvorak HF, Senger DR (1994) Osteopontin
expression and distribution in human carcinomas. Am J Pathol
145(3):61023
Bruemmer D, Collins AR, Noh G, Wang W, Territo M, Arias-
Magallona S, Fishbein MC, Blaschke F, Kintscher U, Graf K,
Law RE, Hsueh WA (2003) Angiotensin II-accelerated athero-
sclerosis and aneurysm formation is attenuated in osteopontin-
deficient mice. J Clin Invest 112(9):13181331
Bulter W (1995) Structural and functional domains of osteopontin.
Ann NY Acad Sci 760:611
Chabas D, Baranzini SE, Mitchell D, Bernard CC, Rittling SR,
Denhardt DT, Sobel RA, Lock C, Karpuj M, Pedotti R, Heller R,
Oksenberg JR, Steinman L (2001) The influence of the
proinflammatory cytokine, osteopontin, on autoimmune demye-
linating disease. Science 294(5547):17311735
Chambers AF, Wilson SM, Kerkvliet N, O'Malley FP, Harris JF,
Casson AG (1996) Osteopontin expression in lung cancer. Lung
Cancer 15(3):31123
Chellaiah MA, Kizer N, Biswas R, Alvarez U, Strauss-Schoenberger
J, Rifas L, Rittling SR, Denhardt DT, Hruska KA (2003)
Osteopontin deficiency produces osteoclast dysfunction due to
reduced CD44 surface expression. Mol Biol Cell 14(1):173189
Chen Y, Bal BS, Gorski JP (1992) Calcium and collagen binding
properties of osteopontin, bone sialoprotein, and bone acidic
glycoprotein-75 from bone. J Biol Chem 267(34):2487124878
Chen J, Singh K, Mukherjee BB, Sodek J (1993) Developmental
expression of osteopontin (OPN) mRNA in rat tissues: evidence
for a role for OPN in bone formation and resorption. Matrix 13
(2):113123
Christensen B, Nielsen MS, Haselmann KF, Petersen TE, Sorensen ES
(2005) Post-translationally modified residues of native human
osteopontin are located in clusters: identification of 36 phosphor-
ylation and five O-glycosylation sites and their biological
implications. Biochem J 390(Pt 1):285292
Comabella M, Pericot I, Goertsches R, Nos C, Castillo M, Blas
Navarro J, Rio J, Montalban X (2005) Plasma osteopontin levels
in multiple sclerosis. J Neuroimmunol 158(12):231239
Crawford HC, Matrisian LM, Liaw L (1998) Distinct roles of
osteopontin in host defense activity and tumor survival during
squamous cell carcinoma progression in vivo. Cancer Res 58
(22):52065215
Dean RA, Overall CM (2007) Proteomics discovery of metalloproteinase
substrates in the cellular context by iTRAQ labeling reveals a diverse
MMP-2 substrate degradome. Mol Cell Proteomics 6(4):611623
Denda S, Reichardt L, Muller U (1998) Identification of osteopontin
as a novel ligand for the integrin alpha8 beta1 and potential role
for this integrin-ligand interaction in kidney morphogenesis.
Molecular biology of the cell 9(6):14251435
Diao H, Kon S, Iwabuchi K, Kimura C, Morimoto J, Ito D, Segawa T,
Maeda M, Hamuro J, Nakayama T, Taniguchi M, Yagita H, Van
Kaer L, Onoe K, Denhardt D, Rittling S, Uede T (2004)
Osteopontin as a mediator of NKT cell function in T cell-
mediated liver diseases. Immunity 21(4):539550
El-Tanani MK, Campbell FC, Kurisetty V, Jin D, McCann M,
Rudland PS (2006) The regulation and role of osteopontin in
malignant transformation and cancer. Cytokine Growth Factor
Rev 17(6):463474
Fan K, Dai J, Wang H, Wei H, Cao Z, Hou S, Qian W, Li B, Zhao J,
Xu H, Yang C, Guo Y (2008) Treatment of collagen-induced
arthritis with an anti-osteopontin monoclonal antibody through
promotion of apoptosis of both murine and human activated T
cells. Arthritis Rheum 58(7):20412052
Fisher LW, Torchia DA, Fohr B, Young MF, Fedarko NS (2001)
Flexible structures of SIBLING proteins, bone sialoprotein, and
osteopontin. Biochem Biophys Res Commun 280(2):460465
Gao YA, Agnihotri R, Vary CP, Liaw L (2004) Expression and
characterization of recombinant osteopontin peptides represent-
ing matrix metalloproteinase proteolytic fragments. Matrix Biol
23(7):457466
Gao C, Guo H, Mi Z, Grusby MJ, Kuo PC (2007) Osteopontin
induces ubiquitin-dependent degradation of STAT1 in RAW264.7
murine macrophages. J Immunol 178(3):18701881
Giachelli CM, Bae N, Almeida M, Denhardt DT, Alpers CE, Schwartz
SM (1993) Osteopontin is elevated during neointima formation in
The role of osteopontin in inflammatory processes 319
rat arteries and is a novel component of human atherosclerotic
plaques. J Clin Invest 92(4):16861696
Giachelli CM, Lombardi D, Johnson RJ, Murry CE, Almeida M
(1998) Evidence for a role of osteopontin in macrophage
infiltration in response to pathological stimuli in vivo. Am J
Pathol 152(2):353358
Golledge J, Muller J, Shephard N, Clancy P, Smallwood L, Moran C,
Dear AE, Palmer LJ, Norman PE (2007) Association between
osteopontin and human abdominal aortic aneurysm. Arterioscler
Thromb Vasc Biol 27(3):655660
Green PM, Ludbrook SB, Miller DD, Horgan CM, Barry ST (2001)
Structural elements of the osteopontin SVVYGLR motif important
for the interaction with alpha(4) integrins. FEBS Lett 503(1):7579
Guo H, Cai CQ, Schroeder RA, Kuo PC (2001) Osteopontin is a
negative feedback regulator of nitric oxide synthesis in murine
macrophages. J Immunol 166(2):10791086
Hamada Y, Nokihara K, Okazaki M, Fujitani W, Matsumoto T,
Matsuo M, Umakoshi Y, Takahashi J, Matsuura N (2003)
Angiogenic activity of osteopontin-derived peptide SVVYGLR.
Biochem Biophys Res Commun 310(1):153157
Hamada Y, Egusa H, Kaneda Y, Hirata I, Kawaguchi N, Hirao T,
Matsumoto T, Yao M, Daito K, Suzuki M, Yatani H, Daito M,
Okazaki M, Matsuura N (2007) Synthetic osteopontin-derived
peptide SVVYGLR can induce neovascularization in artificial
bone marrow scaffold biomaterials. Dent Mater J 26(4):487492
Hu DD, Lin EC, Kovach NL, Hoyer JR, Smith JW (1995) A biochemical
characterization of the binding of osteopontin to integrins alpha v
beta 1 and alpha v beta 5. J Biol Chem 270(44):2623226238
Hur EM, Youssef S, Haws ME, Zhang SY, Sobel RA, Steinman L
(2007) Osteopontin-induced relapse and progression of autoim-
mune brain disease through enhanced survival of activated T
cells. Nat Immunol 8(1):7483
Isoda K, Nishikawa K, Kamezawa Y, Yoshida M, Kusuhara M, Moroi
M, Tada N, Ohsuzu F (2002) Osteopontin plays an important role
in the development of medial thickening and neointimal
formation. Circ Res 91(1):7782
Ito K, Kon S, Nakayama Y, Kurotaki D, Saito Y, Kanayama M,
Kimura C, Diao H, Morimoto J, Matsui Y, Uede T (2009) The
differential amino acid requirement within osteopontin in alpha4
and alpha9 integrin-mediated cell binding and migration. Matrix
Biol 28(1):1119
Jansson M, Panoutsakopoulou V, Baker J, Klein L, Cantor H (2002)
Cutting edge: attenuated experimental autoimmune encephalomy-
elitis in eta-1/osteopontin-deficient mice. J Immunol 168(5):20969
Jono S, Peinado C, Giachelli CM (2000) Phosphorylation of
osteopontin is required for inhibition of vascular smooth muscle
cell calcification. J Biol Chem 275(26):2019720203
Kariuki SN, Moore JG, Kirou KA, Crow MK, Utset TO, Niewold TB
(2009) Age- and gender-specific modulation of serum osteopon-
tin and interferon-alpha by osteopontin genotype in systemic
lupus erythematosus. Genes Immun 10:487494
Katagiri YU, Sleeman J, FujiiH, Herrlich P, Hotta H, Tanaka K, Chikuma
S, Yagita H, Okumura K, Murakami M, Saiki I, Chambers AF, Uede
T (1999) CD44 variants but not CD44s cooperate with beta1-
containing integrins to permit cells to bind to osteopontin indepen-
dently of arginine-glycine-aspartic acid, thereby stimulating cell
motility and chemotaxis. Cancer Res 59(1):219226
Kawamura K, Iyonaga K, Ichiyasu H, Nagano J, Suga M, Sasaki Y (2005)
Differentiation, maturation, and survival of dendritic cells by
osteopontin regulation. Clin Diagn Lab Immunol 12(1):206212
Kazanecki CC, Uzwiak DJ, Denhardt DT (2007) Control of
osteopontin signaling and function by post-translational phos-
phorylation and protein folding. J Cell Biochem 102(4):912924
Koh A, da Silva AP, Bansal AK, Bansal M, Sun C, Lee H, Glogauer
M, Sodek J, Zohar R (2007) Role of osteopontin in neutrophil
function. Immunology 122(4):466475
Kon S, Ikesue M, Kimura C, Aoki M, Nakayama Y, Saito Y, Kurotaki
D, Diao H, Matsui Y, Segawa T, Maeda M, Kojima T, Uede T
(2008) Syndecan-4 protects against osteopontin-mediated acute
hepatic injury by masking functional domains of osteopontin. J
Exp Med 205(1):2533
Krause SW, Rehli M, Kreutz M, Schwarzfischer PJD, Andreesen R
(1996) Differential screening identifies genetic markers of
monocyte to macrophage maturation. J Leukoc Biol 60(4):5405
Lenga Y, Koh A, Perera AS, McCulloch CA, Sodek J, Zohar R (2008)
Osteopontin expression is required for myofibroblast differenti-
ation. Circ Res 102(3):319327
Liaw L, Almeida M, Hart CE, Schwartz SM, Giachelli CM (1994)
Osteopontin promotes vascular cell adhesion and spreading and
is chemotactic for smooth muscle cells in vitro. Circ Res 74
(2):214224
Liaw L, Skinner MP, Raines EW, Ross R, Cheresh DA, Schwartz SM,
Giachelli CM (1995) The adhesive and migratory effects of
osteopontin are mediated via distinct cell surface integrins. Role
of alpha v beta 3 in smooth muscle cell migration to osteopontin
in vitro. J Clin Invest 95(2):713724
Liaw L, Birk DE, Ballas CB, Whitsitt JS, Davidson JM, Hogan BL
(1998) Altered wound healing in mice lacking a functional
osteopontin gene (spp 1). J Clin Invest 101(7):14681478
Maeda K, Takahashi K, Takahashi F, Tamura N, Maeda M, Kon S,
Uede T, Fukuchi Y (2001) Distinct roles of osteopontin frag-
ments in the development of the pulmonary involvement in
sarcoidosis. Lung 179(5):279291
Marcondes MC, Poling M, Watry DD, Hall D, Fox HS (2008) In vivo
osteopontin-induced macrophage accumulation is dependent on
CD44 expression. Cell Immunol 254(1):5662
Marroquin CE, Downey L, Guo H, Kuo PC (2004) Osteopontin
increases CD44 expression and cell adhesion in RAW 264.7
murine leukemia cells. Immunol Lett 95(1):109112
Martin SM, Schwartz JL, Giachelli CM, Ratner BD (2004) Enhancing
the biological activity of immobilized osteopontin using a type-1
collagen affinity coating. J Biomed Mater Res A 70(1):1019
Matsui Y, Rittling SR, Okamoto H, Inobe M, Jia N, Shimizu T, Akino
M, Sugawara T, Morimoto J, Kimura C, Kon S, Denhardt D,
Kitabatake A, Uede T (2003) Osteopontin deficiency attenuates
atherosclerosis in female apolipoprotein E-deficient mice. Arte-
rioscler Thromb Vasc Biol 23(6):10291034
Mi Z, Guo H, Wai PY, Gao C, Kuo PC (2006) Integrin-linked kinase
regulates osteopontin-dependent MMP-2 and uPA expression to
convey metastatic function in murine mammary epithelial cancer
cells. Carcinogenesis 27(6):11341145
Mori R, Shaw T, Martin P (2008) Molecular mechanisms linking wound
inflammation and fibrosis: knockdown of osteopontin leads to rapid
repair and reduced scarring. J Exp Med 205(1):4351
Mukherjee BB, Nemir M, Beninati S, Cordella-Miele E, Singh K,
Chackalaparampil I, Shanmugam V, DeVouge MW, Mukherjee
AB (1995) Interaction of osteopontin with fibronectin and other
extracellular matrix molecules. Ann N Y Acad Sci 760:201212
Murry CE, Giachelli CM, Schwartz SM, Vracko R (1994) Macro-
phages express osteopontin during repair of myocardial necrosis.
Am J Pathol 145(6):14501462
Murugaiyan G, Mittal A, Weiner HL (2008) Increased osteopontin
expression in dendritic cells amplifies IL-17 production by CD4+
T cells in experimental autoimmune encephalomyelitis and in
multiple sclerosis. J Immunol 181(11):74807488
Nagata T, Todescan R, Goldberg HA, Zhang Q, Sodek J (1989)
Sulphation of secreted phosphoprotein I (SPPI, osteopontin) is
associated with mineralized tissue formation. Biochem Biophys
Res Commun 165(1):234240
Nakamachi T, Nomiyama T, Gizard F, Heywood EB, Jones KL, Zhao
Y, Fuentes L, Takebayashi K, Aso Y, Staels B, Inukai T,
Bruemmer D (2007) PPARalpha agonists suppress osteopontin
320 S.A. Lund et al.
expression in macrophages and decrease plasma levels in patients
with type 2 diabetes. Diabetes 56(6):16621670
Nau GJ, Liaw L, Chupp GL, Berman JS, Hogan BL, Young RA
(1999) Attenuated host resistance against Mycobacterium bovis
BCG infection in mice lacking osteopontin. Infect Immun 67
(8):42234230
Nishimichi N, Higashikawa F, Kinoh HH, Tateishi Y, Matsuda H,
Yokosaki Y (2009) Polymeric osteopontin employs integrin
alpha 9beta 1 as a receptor and attracts neutrophils by presenting
a de novo binding site. J Biol Chem 284:1476914776
Nystrom T, Duner P, Hultgardh-Nilsson A (2007) A constitutive
endogenous osteopontin production is important for macrophage
function and differentiation. Exp Cell Res 313(6):11491160
OBrien ER, Garvin MR, Stewart DK, Hinohara T, Simpson JB,
Schwartz SM, Giachelli CM (1994) Osteopontin is synthesized
by macrophage, smooth muscle, and endothelial cells in primary
and restenotic human coronary atherosclerotic plaques. Arterios-
cler Thromb 14(10):16481656
Ogawa D, Stone JF, Takata Y, Blaschke F, Chu VH, Towler DA, Law
RE, Hsueh WA, Bruemmer D (2005) Liver x receptor agonists
inhibit cytokine-induced osteopontin expression in macrophages
through interference with activator protein-1 signaling pathways.
Circ Res 96(7):e5967
Ohri R, Tung E, Rajachar R, Giachelli CM (2005) Mitigation of
ectopic calcification in osteopontin-deficient mice by exogenous
osteopontin. Calcif Tissue Int 76(4):307315
Ohshima S, Yamaguchi N, Nishioka K, Mima T, Ishii T, Umeshita-
Sasai M, Kobayashi H, Shimizu M, Katada Y, Wakitani S,
Murata N, Nomura S, Matsuno H, Katayama R, Kon S, Inobe
M, Uede T, Kawase I, Saeki Y (2002) Enhanced local
production of osteopontin in rheumatoid joints. J Rheumatol
29(10):20612067
Ophascharoensuk V, Giachelli CM, Gordon K, Hughes J, Pichler R,
Brown P, Liaw L, Schmidt R, Shankland SJ, Alpers CE, Couser
WG, Johnson RJ (1999) Obstructive uropathy in the mouse: role
of osteopontin in interstitial fibrosis and apoptosis. Kidney Int 56
(2):571580
ORegan AW, Chupp GL, Lowry JA, Goetschkes M, Mulligan N,
Berman JS (1999) Osteopontin is associated with T cells in
sarcoid granulomas and has T cell adhesive and cytokine-like
properties in vitro. J Immunol 162(2):10241031
ORegan AW, Hayden JM, Berman JS (2000) Osteopontin augments
CD3-mediated interferon-gamma and CD40 ligand expression by
T cells, which results in IL-12 production from peripheral blood
mononuclear cells. J Leukoc Biol 68(4):495502
Panda D, Kundu GC, Lee BI, Peri A, Fohl D, Chackalaparampil I,
Mukherjee BB, Li XD, Mukherjee DC, Seides S, Rosenberg J,
Stark K, Mukherjee AB (1997) Potential roles of osteopontin and
alphaVbeta3 integrin in the development of coronary artery
restenosis after angioplasty. Proc Natl Acad Sci U S A 94
(17):93089313
Panzer U, Thaiss F, Zahner G, Barth P, Reszka M, Reinking R, Wolf
G, Helmchen U, Stahl R (2001) Monocyte chemoattractant
protein-1 and osteopontin differentially regulate monocytes
recruitment in experimental glomerulonephritis. Kidney Int 59
(5):17621769
Patarca R, Freeman GJ, Singh RP, Wei FY, Durfee T, Blattner F,
Regnier DC, Kozak CA, Mock BA, Morse HC 3rd, Jerrells TR,
Cantor H (1989) Structural and functional studies of the early T
lymphocyte activation 1 (Eta-1) gene. Definition of a novel T
cell-dependent response associated with genetic resistance to
bacterial infection. J Exp Med 170(1):145161
Patarca R, Saavedra R, Cantor H (1993) Molecular and cellular basis
of genetic resistance to bacterial infection: the role of the early T-
lymphocyte activation-1/osteopontin gene. Crit Rev Immunol 13
(34):225246
Persy V, Verhulst A, Ysebaert D, De Greef K, De Broe M (2003)
Reduced postischemic macrophage infiltration and interstitial
fibrosis in osteopontin knockout mice. Kidney Int 63(2):543553
Renkl AC, Wussler J, Ahrens T, Thoma K, Kon S, Uede T, Martin SF,
Simon JC, Weiss JM (2005) Osteopontin functionally activates
dendritic cells and induces their differentiation toward a Th1-
polarizing phenotype. Blood 106(3):946955
Rittling SR, Matsumoto HN, McKee MD, Nanci A, An XR, Novick
KE, Kowalski AJ, Noda M, Denhardt DT (1998) Mice lacking
osteopontin show normal development and bone structure but
display altered osteoclast formation in vitro. J Bone Miner Res
13(7):11011111
Rollo EE, Laskin DL, Denhardt DT (1996) Osteopontin inhibits nitric
oxide production and cytotoxicity by activated RAW264.7
macrophages. J Leukoc Biol 60(3):397404
Schulz G, Renkl AC, Seier A, Liaw L, Weiss JM (2008) Regulated
osteopontin expression by dendritic cells decisively affects their
migratory capacity. J Invest Dermatol 128(10):25412544
Senger DR, Perruzzi CA, Papadopoulos-Sergiou A, Van de Water L
(1994) Adhesive properties of osteopontin: regulation by a
naturally occurring thrombin-cleavage in close proximity to the
GRGDS cell-binding domain. Mol Biol Cell 5(5):565574
Senger DR, Perruzzi CA (1996) Cell migration promoted by a potent
GRGDS-containing thrombin-cleavage fragment of osteopontin.
Biochim Biophys Acta 1314(12):1324
Senger DR, Perruzzi CA, Papadopoulos A (1989) Elevated expression
of secreted phosphoprotein I (osteopontin, 2ar) as a consequence
of neoplastic transformation. Anticancer Res 9(5):12911299
Sennels H, Sorensen S, Ostergaard M, Knudsen L, Hansen M, Skjodt
H, Peters N, Colic A, Grau K, Jacobsen S (2008) Circulating
levels of osteopontin, osteoprotegerin, total soluble receptor
activator of nuclear factor-kappa B ligand, and high-sensitivity
C-reactive protein in patients with active rheumatoid arthritis
randomized to etanercept alone or in combination with metho-
trexate. Scand J Rheumatol 37(4):241247
Shinohara ML, Jansson M, Hwang ES, Werneck MB, Glimcher LH,
Cantor H (2005) T-bet-dependent expression of osteopontin
contributes to T cell polarization. Proc Natl Acad Sci U S A
102(47):1710117106
Shinohara ML, Lu L, Bu J, Werneck MB, Kobayashi KS, Glimcher
LH, Cantor H (2006) Osteopontin expression is essential for
interferon-alpha production by plasmacytoid dendritic cells. Nat
Immunol 7(5):498506
Shinohara ML, Kim HJ, Kim JH, Garcia VA, Cantor H (2008a)
Alternative translation of osteopontin generates intracellular
and secreted isoforms that mediate distinct biological activities
in dendritic cells. Proc Natl Acad Sci U S A 105(20):7235
7239
Shinohara ML, Kim JH, Garcia VA, Cantor H (2008b) Engagement of
the type I interferon receptor on dendritic cells inhibits T helper
17 cell development: role of intracellular osteopontin. Immunity
29(1):6878
Sica A, Larghi P, Mancino A, Rubino L, Porta C, Totaro MG, Rimoldi M,
Biswas SK, Allavena P, Mantovani A (2008) Macrophage polariza-
tion in tumour progression. Semin Cancer Biol 18(5):34955
Singh K, DeVouge MW, Mukherjee BB (1990) Physiological
properties and differential glycosylation of phosphorylated and
nonphosphorylated forms of osteopontin secreted by normal rat
kidney cells. J Biol Chem 265(30):1869618701
Smith LL, Giachelli CM (1998) Structural requirements for alpha 9
beta 1-mediated adhesion and migration to thrombin-cleaved
osteopontin. Exp Cell Res 242(1):351360
Smith LL, Cheung HK, Ling LE, Chen J, Sheppard D, Pytela R,
Giachelli CM (1996) Osteopontin N-terminal domain contains a
cryptic adhesive sequence recognized by alpha9beta1 integrin. J
Biol Chem 271(45):2848528491
The role of osteopontin in inflammatory processes 321
Smith LL, Greenfield BW, Aruffo A, Giachelli CM (1999) CD44 is
not an adhesive receptor for osteopontin. J Cell Biochem 73
(1):2030
Sorensen ES, Hojrup P, Petersen TE (1995) Posttranslational
modifications of bovine osteopontin: identification of twenty-
eight phosphorylation and three O-glycosylation sites. Protein Sci
4(10):20402049
Steinman L (2007) A brief history of T(H)17, the first major revision
in the T(H)1/T(H)2 hypothesis of T cell-mediated tissue damage.
Nat Med 13(2):13945
Steitz SA, Speer MY, McKee MD, Liaw L, Almeida M, Yang H,
Giachelli CM (2002) Osteopontin inhibits mineral deposition and
promotes regression of ectopic calcification. Am J Pathol 161
(6):20352046
Strom A, Franzen A, Wangnerud C, Knutsson AK, Heinegard D,
Hultgardh-Nilsson A (2004) Altered vascular remodeling in
osteopontin-deficient atherosclerotic mice. J Vasc Res 41(4):314322
Takafuji V, Forgues M, Unsworth E, Goldsmith P, Wang XW (2007)
An osteopontin fragment is essential for tumor cell invasion in
hepatocellular carcinoma. Oncogene 26(44):63616371
Takahashi K, Takahashi F, Tanabe KK, Takahashi H, Fukuchi Y
(1998) The carboxyl-terminal fragment of osteopontin suppresses
arginine-glycine-asparatic acid-dependent cell adhesion. Bio-
chem Mol Biol Int 46(6):10811092
Tsai AT, Rice J, Scatena M, Liaw L, Ratner BD, Giachelli CM (2005)
The role of osteopontin in foreign body giant cell formation.
Biomaterials 26(29):58355843
Wai PY, Kuo PC (2004) The role of osteopontin in tumor metastasis. J
Surg Res 121(2):228241
Wai PY, Kuo PC (2008) Osteopontin: regulation in tumor metastasis.
Cancer Metastasis Rev 27(1):103118
Wai PY, Guo L, Gao C, Mi Z, Guo H, Kuo PC (2006) Osteopontin
inhibits macrophage nitric oxide synthesis to enhance tumor
proliferation. Surgery 140(2):132140
Weber GF, Ashkar S, Glimcher MJ, Cantor H (1996) Receptor-ligand
interaction between CD44 and osteopontin (Eta-1). Science 271
(5248):509512
Weber GF, Zawaideh S, Hikita S, Kumar VA, Cantor H, Ashkar S
(2002) Phosphorylation-dependent interaction of osteopontin
with its receptors regulates macrophage migration and activation.
J Leukoc Biol 72(4):752761
Weiss JM, Renkl AC, Maier CS, Kimmig M, Liaw L, Ahrens T, Kon
S, Maeda M, Hotta H, Uede T, Simon JC (2001) Osteopontin is
involved in the initiation of cutaneous contact hypersensitivity by
inducing Langerhans and dendritic cell migration to lymph
nodes. J Exp Med 194(9):12191229
Wesson JA, Johnson RJ, Mazzali M, Beshensky AM, Stietz S,
Giachelli C, Liaw L, Alpers CE, Couser WG, Kleinman JG,
Hughes J (2003) Osteopontin is a critical inhibitor of calcium
oxalate crystal formation and retention in renal tubules. J Am Soc
Nephrol 14(1):139147
Xanthou G, Alissafi T, Semitekolou M, Simoes DC, Economidou E,
Gaga M, Lambrecht BN, Lloyd CM, Panoutsakopoulou V (2007)
Osteopontin has a crucial role in allergic airway disease through
regulation of dendritic cell subsets. Nat Med 13(5):570578
Xuan J, Hota C, Chambers A (1994) Recombinant GST-human
osteopontin fusion protein i...[J Cell Biochem. 1994]PubMed
Result. J Cell Biochem 54(2):247255
Yamamoto N, Sakai F, Kon S, Morimoto J, Kimura C, Yamazaki H,
Okazaki I, Seki N, Fujii T, Uede T (2003) Essential role of the
cryptic epitope SLAYGLR within osteopontin in a murine model
of rheumatoid arthritis. J Clin Invest 112(2):181188
Yamamoto N, Nakashima T, Torikai M, Naruse T, Morimoto J, Kon S,
Sakai F, Uede T (2007) Successful treatment of collagen-induced
arthritis in non-human primates by chimeric anti-osteopontin
antibody. Int Immunopharmacol 7(11):14601470
Yokosaki Y, Matsuura N, Sasaki T, Murakami I, Schneider H,
Higashiyama S, Saitoh Y, Yamakido M, Taooka Y, Sheppard D
(1999) The integrin alpha(9)beta(1) binds to a novel recognition
sequence (SVVYGLR) in the thrombin-cleaved amino-terminal
fragment of osteopontin. J Biol Chem 274(51):3632836334
Yokosaki Y, Tanaka K, Higashikawa F, Yamashita K, Eboshida A
(2005) Distinct structural requirements for binding of the
integrins alphavbeta6, alphavbeta3, alphavbeta5, alpha5beta1
and alpha9beta1 to osteopontin. Matrix Biol 24(6):418427
Yu XQ, Nikolic-Paterson DJ, Mu W, Giachelli CM, Atkins RC,
Johnson RJ, Lan HY (1998) A functional role for osteopontin in
experimental crescentic glomerulonephritis in the rat. Proc Assoc
Am Physicians 110(1):5064
Zhao J, Dong L, Lu B, Wu G, Xu D, Chen J, Li K, Tong X, Dai J, Yao
S, Wu M, Guo Y (2008) Down-regulation of osteopontin
suppresses growth and metastasis of hepatocellular carcinoma
via induction of apoptosis. Gastroenterology 135(3):956968
Zhu B, Suzuki K, Goldberg HA, Rittling SR, Denhardt DT,
McCulloch CA, Sodek J (2004) Osteopontin modulates CD44-
dependent chemotaxis of peritoneal macrophages through G-
protein-coupled receptors: evidence of a role for an intracellular
form of osteopontin. J Cell Physiol 198(1):155167
Zohar R, Suzuki N, Suzuki K, Arora P, Glogauer M, McCulloch CA,
Sodek J (2000) Intracellular osteopontin is an integral component
of the CD44-ERM complex involved in cell migration. J Cell
Physiol 184(1):118130
322 S.A. Lund et al.
... It is also secreted in bodily fluids such as blood, milk, and urine (8,9). At sites of inflammation, OPN is secreted from various immune cells, including activated T lymphocytes, macrophages, and leukocytes, and is involved in the pathogenesis of autoimmune and chronic inflammatory disorders (10)(11)(12). Splicing variants of OPN also play a role as tumor promoters and are involved in every step of tumor aggressiveness, including cell proliferation, angiogenesis, and the development of distant metastases. However, the mechanisms by which OPN promotes malignant behavior are still unclear (13). ...
... However, with the current tools, determining the exact source of OPN in patients' serum is impossible. Elevated levels of OPN can lead to changes in the expression of other genes involved in the progression of neoplasms (9)(10)(11)(12). It is important to note that OPN isoforms have distinct expression profiles and biological effects, which may partly explain the diverse expression patterns observed in various types of SGTs, even within a particular cancer type. ...
Article
Background: Osteopontin (OPN) is involved in various biological functions, including cancer pathogenesis and metastasis. However, its clinical value in salivary gland tumors (SGTs) has received little attention. Therefore, this study aimed to evaluate the connection between OPN serum levels and the clinicopathologic features of benign and malignant SGTs in Iranian patients. Methods: This cross-sectional study involved a total of 90 patients with SGTs, including malignant (n=60) and benign (n=30) cases. The control group consisted of 60 healthy individuals matched by age and sex to the patient group. An enzyme-linked immunosorbent assay (ELISA) kit was employed to quantify OPN serum levels in samples. Results: OPN serum levels were significantly elevated in patients compared to the control group (23.16±11.08 ng/mL vs. 10.61±4.99 ng/mL, P<0.0001). There were also significant differences in OPN serum levels between malignant and benign cases of SGT (26.64±11.73 ng/mL vs. 16.19±4.68 ng/mL, P<0.0001). Additionally, OPN concentration was significantly associated with the clinicopathologic characteristics of patients, including tumor size, tumor node metastasis (TNM) classification, and lymph node involvement, but not with patients’ gender, age, and the size and type of lesions. Conclusion: Based on the findings, OPN serum levels could be used as a supportive biomarker to diagnose SGTs and differentiate between benign and malignant cases. This is a preliminary study; therefore, additional research with a larger number of participants and extended monitoring periods is necessary to evaluate the potential of OPN in more effectively diagnosing SGTs in their early stages.
... SERPINE1, an immune-related protein shown to have roles in both cellular senescence (Khan et al., 2017) and cancer , decreased following the 7-day retreat, suggesting that the retreat experience may have antiaging and anti-cancer effects. Finally, OPN, a multi-faceted pro-inflammatory chemokine (Lund et al., 2009), and PGDF-AA, a protein with a role in wound healing and repair, were also reduced in post-meditation plasma. These data indicate trends for both an increase and a decrease in inflammation after the 7-day retreat. ...
Article
Full-text available
Objectives Meditation has long been known to promote health. We utilized a multidisciplinary approach to investigate the impact of mind–body interventions on the body in a twin cohort during a week-long meditation retreat. Method This study was designed to address individual changes controlling for intersubject trait variation and explore the role of genetic background on multi-omic factors during meditation. Transcriptomic analysis was carried out from whole blood samples, while metabolomic and biochemical studies were carried out in blood plasma. Quantitative electroencephalography studies, coupled with biometric analysis and molecular studies at multiple time points, were carried out in twins meditating together and in twins separated and simultaneously either meditating or listening to a documentary. Results Changes in gene expression, metabolites, and cytokines in blood plasma associated with specific meditative states showed patterns of change relative to the time point being assessed. Twin sets were similar in multiple domains before the start of the retreat, showed considerable divergence at the mid-point, and looked more similar by the end of the retreat. Twin pairs showed significant spectral power correlations in separate rooms and when only one twin meditated. These similarities were not observed in mismatched twin pairs. Heart rate dynamics assessments showed alignment among twin pairs, absent between unmatched pairs. Conclusions To our knowledge, this pilot study is novel within the twin research paradigm and is a first step toward exploring the effects of meditation in twins. Preregistration This study was not preregistered and was carried out under IRB protocol MED02#20211477.
... Statistical tests: B: Fisher's exact test with correction for multiple comparisons run in Enrichr. Fig. 13) and binds to many integrins and to CD44 to elicit different cellular functions such as cell migration, cell proliferation, cytokine production, and neurite outgrowth [55][56][57][58][59] . Our interactome data highlighted that osteopontin at Nageotte nodules may be interacting with CD44 found on nearby neurons (Fig. 5A, Supplementary Fig. 12). ...
Article
Full-text available
Nageotte nodules, first described in 1922 by Jean Nageotte, are clusters of non-neuronal cells that form after sensory neuron death. Despite their historical recognition, little is known about their molecular identity nor their involvement in neuropathies that involve neuronal loss like diabetic peripheral neuropathy (DPN). In this study, we molecularly characterize Nageotte nodules in dorsal root ganglia recovered from organ donors with DPN. Here we show that Nageotte nodules are abundant in DPN sensory ganglia and account for 25% of all neurons. Peripherin-and Nav1.7-positive dystrophic axons invade Nageotte nodules, forming small neuroma-like structures. Using histology and spatial sequencing, we demonstrate that Nageotte nodules are mainly composed of satellite glia and non-myelinating Schwann cells that express SPP1 and are intertwined with sprouting sensory axons originating from neighboring neurons. Our findings suggest that Nageotte nodules are an integral feature of dorsal root ganglion neurodegeneration, providing potential therapeutic targets for sensory neuron protection and pain management in DPN.
... But OPN in its role as proinflammatory cytokine and effector on the macrophage response [70] in combination with the activation of the transforming growth factor beta (TGFb) pathway is crucial for nephrolithiasis formation and tubular fibrosis [71][72][73][74][75]. In the trial of Anan et al., they found a large turnover of a variety of inflammation participants as well as macrophage infiltration in the cortical interstitial space in the kidneys of the ethylene glycol-treated rats. ...
Article
Full-text available
Introduction Urolithiasis is one of the most common diseases worldwide, characterized by high morbidity and significant treatment-related costs, with a rising prevalence of up to 20%. The relapse rate within the first 10 years after initial treatment is estimated to be about 60%. Given the increasing prevalence, healthcare-related costs associated with urinary tract stones in the USA are expected to reach up to US $1.24 billion annually by 2030. Current prophylactic therapy for urolithiasis recurrence includes lifestyle modifications, citrate supplementation, and pharmaceuticals. However, a high number of cases remain unresponsive to available pharmacological therapies. Though initially developed for the treatment of Diabetes mellitus, SGLT-2 inhibitors have shown promise in decreasing cardiac and renal endpoints across multiple indications. Recent registry studies have indicated that patients receiving SGLT-2 inhibitors exhibit lower rates of urolithiasis incidence, suggesting a potential reduction in recurrence rates and associated mortality. Objectives We hypothesize that SGLT-2 inhibitors (Dapagliflozin), owing to their multiple pleiotropic effects, may offer a viable treatment option for the prophylaxis of high-risk calcium oxalate kidney stones and reduce urinary calcium oxalate output. Methods This study will proceed in two phases: an exploratory phase and a randomized controlled phase. In the exploratory phase, 22 participants with indications for dapagliflozin treatment will be evaluated before and after treatment initiation to ascertain the concrete effect size regarding oxalate and calcium-sparing effects. This data will inform the calculation of the study sample size (ranging from 17 to 104 participants) to include high-risk calcium oxalate kidney stone formers in a randomized controlled crossover study design. Treatment phases—one with dapagliflozin and one with placebo—will alternate with wash-out phases involving placebo. The primary outcome is the reduction of oxalate excretion in 24-hour urine samples compared to baseline values after 8 weeks of therapy. Secondary objectives include analysing effects on kidney function, the frequency of urolithiasis, and treatment tolerance. Additionally, in-depth metabolomics analyses will explore pathophysiological pathways during treatment. Investigators, patients, and research staff will be blinded to the randomization list. This study was initially registered under EudraCT (Nr:2022-000994-13) and has been transitioned to CTIS (Nr: 2024-519371-25-00) to comply with EU Regulation 536/2014, ensuring streamlined management and transparency. Discussion Dapagliflozin’s pleiotropic effects may provide a novel prophylactic treatment option for urolithiasis. This study aims to evaluate potential treatment effects in a prospective RCT and elucidate potential pathophysiological pathways through in-depth metabolomics analyses. SGLT-2 inhibitors have the potential to transform the landscape of urolithiasis treatment, reduce the healthcare burden on individuals and the system, and significantly improve patient quality of life.
... Osteopontin (OPN), a secreted extracellular matrix protein, that interacts with Integrins, functions as a Th1 cytokine, is involved in tissue remodeling (Lund et al. 2009) and intricately linked to the JAK2/STAT3 and PI3K/Akt signaling pathways in HCC, contributing to tumor growth, invasion, and metastasis (Desert et al. 2022;Wu et al. 2022;Yu et al. 2018). Studies also found that increased secretion of OPN contributed to promoting the synthesis of collagen-I in hepatic stellate cells via inducing HMGB1 (Arriazu et al. 2017), which is involved in chronic liver disease (Song et al. 2021). ...
Article
Full-text available
Hepatocellular carcinoma (HCC) is the third major cause of cancer death worldwide, with more than a doubling of incidence over the past two decades in the United States. Yet, the survival rate remains less than 20%, often due to late diagnosis at advanced stages. Current HCC screening approaches are serum alpha-fetoprotein (AFP) testing and ultrasound (US) of cirrhotic patients. However, these remain suboptimal, particularly in the setting of underlying obesity and metabolic dysfunction-associated steatotic liver disease/steatohepatitis (MASLD/MASH), which are also rising in incidence. Therefore, there is an urgent need for novel biomarkers that can stratify risk and predict early diagnosis of HCC, which is curable. Advances in liver cancer biology, multi-omics technologies, artificial intelligence, and precision algorithms have facilitated the development of promising candidates, with several emerging from completed phase 2 and 3 clinical trials. This review highlights the performance of these novel biomarkers and algorithms from a mechanistic perspective and provides new insight into how pathological processes can be detected through blood-based biomarkers. Through human studies compiled with animal models and mechanistic insight in pathways such as the TGF-β pathway, the biological progression from chronic liver disease to cirrhosis and HCC can be delineated. This integrated approach with new biomarkers merit further validation to refine HCC screening and improve early detection and risk stratification.
... This is due to the OPN alteration on the biological function of mesangial cells and podocytes through the secretion of TGF-β and MCP-1 [11]. In addition, increased OPN levels also correlate with increased chronic inflammation in patients with CKD [44]. This is in line with previous findings, which show that OPN levels are directly proportional to inflammatory markers such as CRP and IL-6 [45]. ...
Article
Full-text available
Background The global prevalence of chronic kidney disease (CKD) as a major renal disease is increasing rapidly. The progression of CKD may lead to end-stage renal disease (ESRD). Current diagnostic and prognostic methods still have some limitations. This study aims to evaluate the potential and predictive ability of Osteopontin (OPN), Kidney injury molecule-1 (KIM-1), and Fetuin-A on the incidence of ESRD in CKD patients. Methods A systematic review and meta-analysis were carried out based on the PRISMA guideline on registered databases for studies published up to December 21, 2023. The concentrations of each marker were then reported in pooled standardized mean difference (SMD) and hazard ratio (HR). Subgroup analysis was performed based on age, location, and KIM-1 specimen. Results We included 21 studies involving 15,983 patients. Meta-analysis revealed that increasing OPN (SMD = 5.52, 95% CI = 1.59–9.44, p = 0.01) and KIM-1 (SMD = 1.45, 95% CI = 0.50–2.39, p = 0.0027), as well as decreasing Fetuin-A level (SMD = -1.31, 95% CI = -2.37 – -0.26, p = 0.01) were significant in CKD patients with ESRD. Chronic kidney disease patients with increased KIM-1 levels showed 1.13 times increased risk of ESRD (HR = 1.13, 95% CI = 1.10–1.17, p <0.0001). Subgroup analysis showed that increased KIM-1 in urine or blood was strongly associated with ESRD, and decreased Fetuin-A levels in Asians had a significant association with the incidence of ESRD. Conclusion Osteopontin, KIM-1, and Fetuin-A significantly reflect ESRD in CKD patients, making them potential prognostic indicators.
Article
Extracellular matrix remodeling is a natural response to injury but, excessive extracellular matrix accumulation, or fibrosis, is a causative factor in hundreds of diseases that limit organ function, regenerative responses, and can interfere with regenerative therapies. Fibrosis is closely related to inflammation, both of which occur in the salivary glands of patients treated with radiation for head and neck cancers and in patients suffering from autoimmune conditions, such as Sjögren's Disease. Despite the known involvement of fibrosis in disease and the inhibitory effects of fibrosis on tissue regeneration, the mechanisms through which extracellular matrix is elaborated in the salivary gland are poorly understood. Stromal fibroblasts are the primary matrix-producing cells and are known to drive both fibrosis and inflammation. To define the temporal responses of fibroblasts to injury, we induced a temporary obstructive injury though ligation of the primary submandibular and sublingual salivary gland ducts and then performed single-cell RNA sequencing and pathway analysis at timepoints immediately following the injury. Using bioinformatic approaches, we identified three unique fibroblast groups that dynamically respond to the injury. We characterized the changes in matrisomal and inflammatory gene expression over a 7-day time course and identified one group of fibroblasts to be the primary injury-responsive fibrogenic cell type. Understanding how fibroblasts respond at the early and later injury timepoints, along with defining signaling pathways regulated by fibroblasts, could lead to a better understanding of the contribution of fibroblast to acute injury responses to facilitate the development of therapeutics that minimize fibrosis and promote regenerative gland responses in chronic disease states.
Article
Introduction Approximately 39 million people live with HIV globally, with 1.3 million new infections annually. Despite improved treatment, noncommunicable diseases (NCDs) such as cardiovascular disease (CVD), neurological disorders, chronic kidney disease (CKD), and cancer are now the leading causes of death among people with HIV (PWH). Osteopontin (OPN) has emerged as a notable mediator in the inflammatory response to HIV and related NCDs. Our aim is to review the current understanding of OPN's role in HIV-related inflammatory pathways to highlight potential therapeutic avenues for improved treatment and mitigation of comorbidities. Methods We conducted a systematic review by searching relevant literature using specific keywords related to HIV, osteopontin, cardiovascular disease, inflammation, neurological disorders, cancer, and chronic kidney disease. The collected studies were organized and categorized by key themes, followed by a comprehensive analysis to identify patterns and draw conclusions regarding OPN's role in HIV-associated comorbidities. Results The intricate interactions between OPN, its isoforms, and HIV-related illnesses suggest that OPN can exhibit both pro-inflammatory and anti-inflammatory roles, depending on the stage of the disease and the specific cell type involved. Its functions are diverse throughout the progression of HIV and its associated comorbidities, including CVD, CKD, cancer, and neurological disorders. Conclusion OPN's effects on the disease progression of HIV and related NCDs are highly variable due to its diverse functions. Therefore, further research is essential to fully understand its complex roles before considering OPN as a therapeutic target for HIV and its comorbidities.
Article
Neutrophil-independent macrophage responses are a prominent part of delayed-type immune and healing processes and depend on T cell-secreted cytokines. An important mediator in this setting is the phosphoprotein osteopontin, whose secretion by activated T cells confers resistance to infection by several intracellular pathogens through recruitment and activation of macrophages. Here, we analyze the structural basis of this activity following cleavage of the phosphoprotein by thrombin into two fragments. An interaction between the C-terminal domain of osteopontin and the receptor CD44 induces macrophage chemotaxis, and engagement of β3-integrin receptors by a nonoverlapping N-terminal osteopontin domain induces cell spreading and subsequent activation. Serine phosphorylation of the osteopontin molecule on specific sites is required for functional interaction with integrin but not CD44 receptors. Thus, in addition to regulation of intracellular enzymes and substrates, phosphorylation also regulates the biological activity of secreted cytokines. These data, taken as a whole, indicate that the activities of distinct osteopontin domains are required to coordinate macrophage migration and activation and may bear on incompletely understood mechanisms of delayed-type hypersensitivity, wound healing, and granulomatous disease.
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
Osteopontin (OPN) is an extracellular matrix protein that binds to integrin alpha(v) beta(3), Here we demonstrate that two other integrins, alpha(v) beta(1) and alpha(v) beta(5), are also receptors for OPN. Human embryonic kidney 293 cells adhere to human recombinant osteopontin (glutathione S-transferase-osteopontin; GST-OPN) using integrin alpha(v) beta(5). When the 293 cells are transfected with the beta(5) subunit, they can also adhere to GST-OPN using integrin alpha(v) beta(5). Divalent cations regulate the binding of GST-OPN to both alpha(v) beta(1) and alpha(v) beta(5). Mg2+ and Mn2+ support the binding of GST-OPN to these integrins but Ca2+ does not, The highest affinity is observed in Mn2+, In the presence of this ion, the affinity of GST-OPN for alpha(v) beta(1), is 18 nM and the affinity for gp, is 48 nM, The antibody 8A2, which is an agonist for beta(1), promotes the adhesion of 293 cells to GST-OPN even when Ca2+ is present. This observation suggests that cellular events could modulate the affinity of alpha(v) beta(1) for OPN, Collectively, these findings prove that integrins alpha(v) beta(1), alpha(v) beta(3) and alpha(v) beta(5) have similar affinity for OPN. Therefore, all three integrins must be considered when evaluating the biological affects of OPN.
Article
Osteopontin is an adhesive glycoprotein implicated in numerous diseases associated with inflammation and remodeling. There are several structural domains in osteopontin which are of particular interest. The RG1) motif is a cell-attachment sequence shown to be critical for cell adhesion through ovconiaining integrins. In close proximity to the RGD domain is the thrombin cleavage site. Previous observations suggest that thrombin cleavage of osteopuntin occurs in vivo and may be physiologically important. To study the functional significance of osteopontin cleavage by thrombin. we made glutathione-S-transferase-osteopontin fusion proteins. These proteins contain either the N- or C-terminal domains expected to be formed following thrombin cleavage at the Arg\Q9)-Sr 170) peptide bond. We compared these osteopontin fragments with native osteopontin in their ability to support adhesion of several differpnt cell lines, and identified the receptors mediating these interactions. Our data show that the N-terminal osteopontin fragment, which contains the RGD domain, supports adhesion of a melanoma cell line unable to bind native osteopontin. This suggests that osteopontin adhesive interactions may be regulated by thrombin cleavage. We also demonstrate that thrombin cleavage of osteopontin exposes a cryptic adhesive domain which can be recognized by a novel osteopontin receptor. This receptor has been identified as the a98l integrin. This work was supported by NIH training grant 5T32 GM07270-2Ü and NIH grant HL-18645.
Article
For over 35 years, immunologists have divided T-helper (TH) cells into functional subsets. T-helper type 1 (TH1) cells - long thought to mediate tissue damage - might be involved in the initiation of damage, but they do not sustain or play a decisive role in many commonly studied models of autoimmunity, allergy and microbial immunity. A major role for the cytokine interleukin-17 (IL-17) has now been described in various models of immune-mediated tissue injury, including organ-specific autoimmunity in the brain, heart, synovium and intestines, allergic disorders of the lung and skin, and microbial infections of the intestines and the nervous system. A pathway named TH17 is now credited for causing and sustaining tissue damage in these diverse situations. The TH1 pathway antagonizes the T H17 pathway in an intricate fashion. The evolution of our understanding of the TH17 pathway illuminates a shift in immunologists' perspectives regarding the basis of tissue damage, where for over 20 years the role of TH1 cells was considered paramount.
Article
The osteopontin SVVYGLR motif binds the integrins α4β1 and α9β1. We show that α4β7 also interacts with this motif and that an SVVYGLR-OH peptide antagonises the α4β7 MAdCAM interaction. The important elements of this motif required to bind α4β1 and α4β7 were probed using a series of mutated peptides based around SVVYGLR. Leu167 is important for the interaction with α4 integrins, as is the C-terminal carboxylic acid of Arg168 exposed by thrombin cleavage. The importance of the acidic group means that SVVYGLR has structural elements in common with other α4 integrin-binding motifs and suggests why thrombin cleavage activates this motif.