You are on page 1of 32

Accepted Manuscript

The multiple functions and mechanisms of osteopontin

Mehmet Arif İçer, Makbule Gezmen-Karadağ

PII: S0009-9120(18)30383-7
DOI: doi:10.1016/j.clinbiochem.2018.07.003
Reference: CLB 9817
To appear in: Clinical Biochemistry
Received date: 12 April 2018
Revised date: 3 July 2018
Accepted date: 8 July 2018

Please cite this article as: Mehmet Arif İçer, Makbule Gezmen-Karadağ , The multiple
functions and mechanisms of osteopontin. Clb (2018), doi:10.1016/
j.clinbiochem.2018.07.003

This is a PDF file of an unedited manuscript that has been accepted for publication. As
a service to our customers we are providing this early version of the manuscript. The
manuscript will undergo copyediting, typesetting, and review of the resulting proof before
it is published in its final form. Please note that during the production process errors may
be discovered which could affect the content, and all legal disclaimers that apply to the
journal pertain.
ACCEPTED MANUSCRIPT

The Multiple Functions and Mechanisms of Osteopontin

Mehmet Arif İçer1, Makbule Gezmen-Karadağ1

1
Gazi University, Faculty of Health Sciences, Nutrition and Dietetics Department, 06500

Beşevler /ANKARA, TURKEY

PT
Corresponding Author: Mehmet Arif İÇER, Research Assistant, Gazi University, Faculty of
Health Sciences, Nutrition and Dietetics Department, Ankara/Turkey, e-mail:

RI
m.arif.icer@gmail.com, GSM: +905074759248

SC
NU
MA
E D
PT
CE
AC
ACCEPTED MANUSCRIPT

Abstract

Osteopontin (OPN) is a highly phosphorylated glycophosphoprotein having acidic

characteristics and rich in aspartic acid. OPN, a multifunctional protein, has important

functions on cardiovascular diseases, cancer, diabetes and kidney stone diseases and in the

process of inflammation, biomineralization, cell viability and wound healing. Osteopontin

PT
acts on organisms by playing a key role in secretion levels of interleukin-10 (IL-10),

interleukin-12 (IL-12), interleukin-3 (IL–3), interferon-γ (IFN-γ), integrin αvB3, nuclear

RI
factor kappa B (NF-kB), macrophage and T cells, regulating the osteoclast function and

SC
affecting CD44 receptors. The aim of the present review is to address majority of different

functions of OPN protein which are known, suspected or suggested through the data obtained
NU
about this protein yet.
MA

Key words: Osteopontin, Biomarker, Medicinal biochemistry, Obesity, Cancer


E D
PT
CE
AC
ACCEPTED MANUSCRIPT

1. Introduction

Osteopontin (OPN), which is a combination of the words "osteo" meaning "bone" and

"bridge" meaning "pontin", has important roles in health [1]. OPN, the first extracellular

matrix protein identified in bone tissue by Franzén et al., is a highly phosphorylated

glycoprotein consisting of approximately 314 amino acids with a molecular weight ranging

PT
between 44 and 75 kDa. [2, 3].

RI
Osteopontin exists in a large number of tissues such as saliva, milk, bile, dentin layer of the

SC
tooth, kidney, brain, bone marrow, endothelial cell, smooth muscle cells, skeletal muscle cell,

mammary gland cells, chorionic villus, decidua layer of the uterus, the ganglia in inner ear,
NU
salivary and sudoriferous glands, bile and pancreatic tracts [4, 5]. Furthermore, OPN in the

osseous tissue is released from osteoblasts and osteoclasts acting in bone structuring.
MA

However, OPN expressed from osteoclasts is known to inhibit the hydroxyapatite [4] which

indicate that OPN is associated with bone destruction [6].


E D

Different studies have shown that OPN increases macrophages and T cells in the event of
PT

inflammation [4, 7]. Furthermore, there are studies suggesting that OPN suppress ectopic
CE

calcification and over-mineralization, and takes active roles in wound healing and preventing

renal stone formation [4, 8-10]. However, along with many studies indicating positive effects
AC

of OPN, some studies address negative effects of OPN such as increasing OPN release during

tumor development and metastasis and playing a key role in development of insulin resistance

by inducing the inflammation formation [11-13].

Some studies report that release of OPN is affected by some dietary factors [14, 15]. Some of

the aforesaid studies report that some foods increase OPN secretion whereas others suggest

that some foods reduce OPN secretion [14, 16].


ACCEPTED MANUSCRIPT

In the present review, OPN was examined within a wide perspective from structure and

release of OPN, positive health effects to association of negative health effects.

2. Structure and Release of Osteoprotein

Osteopontin is a highly phosphorylated glycophosphoprotein which is rich in aspartic acid

and has acidic characteristics consisting of 300 amino acids and including O-linked and N-

PT
linked oligosaccharides [17]. Structural form of OPN is shown in Figure 1.

RI
It is accepted that OPN has an arginine-glycine-aspartic acid binding zone, 2 heparin binding

SC
zones, one thrombin binding zone and one calcium binding zone. Moreover, matrix

metalloprotease 3 and 7 (MMP-3,7) are also linked to OPN. Osteopontin has two terminal
NU
zones including N-terminal and C-terminal. C-terminal binds two heparin molecules as well
MA

as CD44 variants whereas N-terminal includes integrin receptor binding zones [18].

Although modulation mechanism of OPN release is not fully clarified yet, release levels are
D

reported to be different in different cell types [4, 19]. Osteopontin release is modulated by
E

many factors through different signal pathways including protein kinase C (PKC) on certain
PT

cell types. For instance, PKC suppress OPN release in Src -/- fibroblasts stimulated by
CE

epidermal growth factor [20, 21]. Moreover, increased OPN release in case of injury and

infection is associated with cytokines [20].


AC

Previous studies report that vitamin D, interferon stimulating elements, glucocorticoids and

alignment rich in purine may stimulate OPN release [4, 19]. Moreover, macrophages, tumor

necrosis factor-α (TNFα) and interleukin-1β (IL-1β) stimulate the transcription and release of

OPN genes [4, 19]. Coppola et al. detected in a study at cellular level that OPN release

increased in tumor cells [22].


ACCEPTED MANUSCRIPT
Thrombine Binding Zone

PT
MMP Binding Zone

RI
Figure 1. Structural form of osteopontin [17].

SC
3. Effects of Osteopontin on Health NU
Osteopontin takes important roles in inflammation, biomineralization, cardiovascular

diseases, cellular viability, cancer, diabetes and renal stone disease through different
MA

mechanisms (Figure 2).


D

Biomineralization
Inflammation
Modulation of
E

Increase in macrophage
osteoclast function
and T cell count
PT
CE

Wound healing Cancer


Increase in macrophage Increase in cell
infiltration and TGF- β OSTEOPONTIN
migration
secretion
AC

Nephrolithiasis
Diabetes Prevention of Caox
crystal formation and
Increase in insulin
accumulation
resistance

Figure 2. Major biological functions of osteopontin [2, 8, 12, 13, 23, 24].
ACCEPTED MANUSCRIPT

3.1. Osteopontin and inflammation

Inflammation is a response to prevent damage of microorganisms or toxins at cellular level or

to protect the organism as a result of any injury [25]. The roles of OPN on immune cells

during inflammation and apoptosis processes were shown in Figure 3. OPN is a significant

chemical attractant for macrophages and T cells and many researches were carried out on role

PT
of this substance during inflammation [4, 7, 26]. An increase in OPN release is reported from

macrophages, activated T cells, epithelial and endothelial cells as well as smooth muscle cells

RI
during inflammation process. However, OPN takes a role in increase of macrophage and T

SC
cells on inflammation areas [4, 7]. Moreover, obesity which is characterized by low grade

inflammation in adipose tissue, abnormal production of cytokines and macrophage infiltration


NU
is reported to increase OPN concentration both in the plasma and adipose tissue [27-29].
MA

There are many studies focusing on the roles of OPN glycoproteins during inflammation

process.
D

It was reported that severity of herpex simplex, Mycobacterium bovis BCG and listeria
E

monocytogenes-induced infections increase in tissues of OPN-null mice [7, 26]. A study


PT

conducted on human found out that osteopontin gene expression in human pulmonary
CE

macrophages increase after infection with virulent Mycobacterium bovis BCG [30]. This case

is associated with direct suppression of macrophage production through increased interleukin-


AC

10 (IL-10), decreased interleukin-12 (IL-12), interleukin-3 (IL–3) and interferon-γ (IFN-γ) [7,

26]. Another study conducted on humans found out that OPN resisted myobacterial infections

and diseases [31]. Isumi et al. (2015) evaluated serum OPN levels in the patients with

rheumatoid arthritis and detected statistically significant and lower OPN levels after a 1-year

treatment carried out successfully when compared with the period before treatment [32].

Schaller et al. (2009) evaluated plasma OPN levels of 31 obese patients before and after
ACCEPTED MANUSCRIPT

development of inflammation concomitant to bariatric surgery and concluded that the

postoperative inflammation causes an increase in plasma OPN levels [29]. However, possible

absorption disorders following a major procedure like bariatric surgery may affect plasma

OPN concentrations.

It may be considered due to the aforesaid roles that OPN may increase the inflammation in

PT
acute and chronic inflammatory diseases [4, 26, 31].

RI
SC
NU
MA
E D
PT
CE
AC

Figure 3. Role of osteopontin on immune cells during inflammation and apoptosis [33].

3.2 Osteopontin and biomineralization

As mentioned before, OPN is released in mineralized tissues including bones and teeth and

produced by osteoclastic and osteoblastic cells at high levels [20, 34]. Osteopontin synthesis

stimulation is performed by calcitriol which is known as trigger of bone destruction [35].


ACCEPTED MANUSCRIPT

Furthermore, OPN which is one of the most strongly linked non-collagen proteins is

considered to assist binding of the osteoclasts onto the bone surface [35, 36]. Due to the

effects mentioned above, OPN has 3 major functions during biomineralization phase

including modulation of adherence of osseous cells, modulation of osteoclastic function and

modulation of matrix mineralization.

PT
Osteopontin increases the adhesion of bone cells by concentrating in mineralized collagen

matrix during formation of the bone tissue [8, 37]. Furthermore, OPN increases adhesion of

RI
osteoblast and osteoclast cells [8, 20]. However, there was not any kind of abnormality on

SC
bone and tooth development processes of the rats in the studies conducted on the OPN-null
NU
mutant rats [38, 39]. These studies pointed out that other bone matrix proteins such as osseous

sialoprotein or fibronection would perform the adherence and signal functions required for
MA

osseous cells [8].

Osteopontin takes a role in bone tissue destruction by two basic mechanisms. The first one is
D

providing identification and adherence of osteoclastic cells by integrin αvβ3-mediated OPN;


E
PT

and the second is modulation of osteoclastic function via integrin αvβ3 [35, 36, 40].

Moreover, it is reported that OPN acts as a physical limiter limiting crystal formation in bones
CE

and teeth [8].


AC

In line with such data, OPN which has a significant importance [8, 9] in suppression of over-

mineralization may be considered to reduce the risk of bone fractures through the aforesaid

mechanisms.
ACCEPTED MANUSCRIPT

3.3 Osteopontin and cardiovascular diseases

It is stated that OPN, identified for the first time by Gihemlli et al. in the arterial tissue, may

play significant roles in development of atherosclerosis [41, 42]. Although the mechanisms of

OPN related to cardiovascular diseases are not clear, different possible mechanisms exist. One

of these possible mechanisms is OPN increasing the risk of atherosclerosis by increasing

PT
endothelial cell migration via αvβ3 ligand. Other possible mechanisms include OPN leading

vascular inflammation by increasing macrophage activation and release of other cytokines or

RI
leading development of atherosclerosis by increasing calcification in arteries [42, 43].

SC
Glucose-dependent insulinotropic polypeptide (GIP) which is including effects on the
NU
cardiovascular system is the main incretin hormones secreted by the intestine after a meal to

stimulate insulin secretion [44]. It is also reported that GIP promotes OPN expression in
MA

pancreatic β-cells and thus has a proliferative and antiapoptotic role in this tissue [45].

Interestingly, a single nucleotide polymorphism (SNP) (rs1800437) in the GIPR gene has
E D

been associated with the features of CVD [46]. In a study conducted by Berglund et al. (2015)
PT

a strong association between OPN and GIP hormone was found in patients with advanced

atherosclerotic disease [47]. It is also stated that incretin hormone GIP may increase
CE

cardiovascular disease risk by increasing OPN release in arteries [45, 47, 48].
AC

Vascular calcification, one of the most significant signs of atherosclerosis, is a common

condition detected in the patients with coronary artery diseases. Furthermore, vascular

calcification is known to be associated with increased risk for myocardial infarction [49].

Many studies state that OPN increases risk of atherosclerotic plaque formation leading to

calcification in arteries [50, 51].


ACCEPTED MANUSCRIPT

Dimitrow et al. (2011) implemented atorvastatin treatment (20 mg/day) on 33 individuals with

heart disease for four weeks and examined the effect of atorvastatin treatment on calcification

biomarkers. A statistically significant decrease in OPN levels as a calcification biomarker was

detected following atorvastatin treatment (p<0,05) [52]. In a study where the researchers

evaluated the mineralization developed on smooth muscles of cattle aorta, arterial

calcification was associated with release of OPN [42]. Abdalrhim et al. (2016) concluded in

PT
their study that increased plasma levels of OPN which has important roles in calcifications

RI
and inflammatory processes is associated with coronary artery disease [51]. Another study

SC
found higher OPN levels in cardiac valves with calcification when compared with not-

calcified cardiac valves [53]. In accordance with these data, it may be considered to use OPN
NU
level to detect existence and severity of coronary artery diseases.
MA

3.4. Osteopontin and wound healing-cell vitality

Osteopontin plays important roles in wound healing due to its effects on cell viability. Also
E D

some studies suggest that OPN takes these roles through different possible mechanisms. One
PT

of these mechanisms is protection of endothelial cells by OPN against continuous apoptosis

by increasing the nuclear factor kappa B (NF-kB) via binding to integrin αvB3 [4]. Another
CE

possible mechanism is active role of OPN stimulating IL-3 (Interleukin-3) and GM-CSF
AC

(granulocyte-macrophage colony-stimulating factor) receptors on cell vitality by binding to

CD44 which is a cell surface receptor. In addition, it is known that OPN increase release of

osteoprotegenin (OPG) by increasing NF-kB activation in endothelial cells [54].

Osteoprotegenin protein is known as a suppressor of apoptosis [54, 55]. Along with the

effects on cell vitality, OPN is suggested to have important roles in wound healing by

modulating secretion of metalloproteinase (MMP) and leveraging the fibroblast proliferation

dependent to the growth factor [4]. Furthermore, healing effect of OPN secreted with high
ACCEPTED MANUSCRIPT

levels by tissue macrophages is reported by affecting differentiation and mineralization of

osteoblasts [56]. The studies evaluating effects of OPN on wound healing were generally

carried out on the rats and focused on its effects on cell viability.

Lin et al. (2000) concluded in their study conducted at cell level that OPN increase the cell

vitality level by binding to CD44 receptor [57]. In another study performed on rats with renal

PT
damage, effect of OPN level was evaluated on cell vitality in kidney tissue. As a result, the

rats with lower OPN levels were found to have less cell vitality [23]. Miyazaki et al. (2008)

RI
evaluated the effects of OPN on corneal wound healing through comparison of the OPN-

SC
removed rats with the control group. It was concluded in the aforesaid study that decreased

release of myofibroblasts as well as growth factor β1 cause a delay in wound healing in OPN-
NU
removed rats when compared with the control group. More experimental studies are needed
MA

to clarify the mechanism of action of OPN, which is involved in the regeneration, release and

differentiation of fibroblasts and myofibroblasts, as well as their cell viability and wound
D

healing [58].
E
PT

3.5. Osteopontin and cancer


CE

OPN produced by many species including human being is a multi-functional protein released

by many cells such as tumor cells and macrophages [12]. There are different studies
AC

conducted on humans and animals reporting that release of OPN increases in tumor

development and metastases [59, 60]. In majority of these studies, increase in OPN release in

breast, gastric, lung, prostate, liver and colon cancers is associated with tumor development,

progression and/or metastasis [12, 59, 60]. In line with these studies, use of OPN level

analysis is considered to measure metastasis potential of some tumors [11, 12].


ACCEPTED MANUSCRIPT

Osteopontin takes an important role in tumor development by leading to cell migration in

extracellular matrix [11]. However, OPN is considered to cause tumor progression by

affecting the receptors of different integrin and CD44 [11]. Furthermore, it is reported that

increase in OPN release progresses parallel to severity of helicobacter pylori infection, which

is one of the most important causes for gastric cancer [61, 62]. The role of OPN in gastric

cancer formation has not been explored as yet. Considering OPN to enhance the cell survival

PT
and proliferation, it may also play important roles in the helicobacter pylori-related

RI
precancerous changes [62]. Another possible mechanism is that increased expression of BCR-

SC
ABL1 gene increases OPN release in case of chronic myeloid leukemia [63]. Many other

studies focus on the correlation between OPN secretion level and development, progression
NU
and/or metastasis of tumor in different cancer types.
MA

Many studies report that OPN expression takes important roles in different possible

mechanisms of action in different cancer types (Table 1). Chang et al. (2011) performed a
D

study on 105 patients infected with helicobacter pylori (42 patients with intestinal metaplasia
E

(IM) and 63 patients without IM) and 29 healthy individuals; and found out that gastric OPN
PT

release of the patient group was statistically and significantly higher than the healthy group
CE

[62]. Different studies suggested that increase in OPN release is effective on prognosis of the

gastric cancer through phosphoinositide 3-kinase (PI3K)/AKT pathway, activator of


AC

transcription 1 (STAT1) and pro-inflammatory immune response [64, 65]. Chagan-Yasutan et

al. (2011) reported a statistically significant positive correlation between plasma OPN and

CD44 levels and the disease severity in their study carried out on 27 adults with T cell

leukemia and 30 healthy individuals [66]. Furthermore, it was detected that expression of

OPN increase in acute myeloid leukemia and chronic myeloid leukemia [63, 67]. Psyrri et al.

performed a retrospective study on 1681 patients with early stage breast cancer and showed a

negative correlation between survival rates of the individuals with breast cancer and high
ACCEPTED MANUSCRIPT

OPN mRNA expression. Moreover, the aforesaid study concluded that OPN might be used as

a biomarker in breast cancer [68].

In consideration of the study results, release of OPN which increases in some cancer types

may be used as a clinical biological marker for diagnosis and monitoring of cancer [11, 12,

60]. However, the molecular mechanism defining the role of OPN has not been clarified yet

PT
and it is necessary to further clarify multifactorial mechanisms of action of OPN so that it can

be used for cancer follow-up and diagnosis [12].

RI
SC
NU
MA
E D
PT
CE
AC
ACCEPTED MANUSCRIPT

Table 1 Expression of osteopontin in different cancer types

Cancerous Method Pathophysiological Mechanism Reference


tissue
Ostepontin, E-cadherin, b-catenin and Highly release of OPN appears by the
Gastric cyclooxygenase 2 expression analyses gastric tumor tissue. It was reported [69]
were performed on 346 gastric tumor that highly released OPN affect
tissues by immunohistochemistry adherence and migration of cancer
method. cells.

Plasma OPN levels were measured in A statistically significant positive

PT
T cell 27 adults with T cell leukemia and 30 correlation was reported between [66]
healthy individuals. plasma OPN and CD44 levels and
disease severity.

RI
Prostate biopsy was performed on 40 OPN was over expressed in prostate
Prostate patients with prostate cancer and 30 cancer. This was considered that OPN [70]

SC
patients with benign prostate may be used as an important biomarker
hyperplasia; and anti-OPN antibodies for diagnosis and prognosis of prostate
were analyzed through cancer.
immunohistochemical staining test.
NU
Furthermore, serum levels of OPN
isoforms and total prostate specific
antigen were measured.
MA

OPN concentrations as well as Lower plasma OPN levels were found


Lung plasminogen activator inhibitor-1 (PAI- associated with recovered clinical [71]
1) and vascular endothelial growth outcomes in treatment of the llung
factor (VEGF) concentrations were cancer. Survival rates of the patients
D

measured through enzyme-linked with lower OPN levels are higher in


immunosorbent assay. general.
E

Serum and plasma samples were The patients presenting metastases


PT

Rectal obtained from the patients with rectal have higher OPN release levels both [72]
carcinoma carcinoma before and after before and after chemotherapy when
chemotherapy; and some analyses were compared with the patients without
CE

executed through IL-6 ELISA kit, metastasis. Detection of OPN levels


ostepontin ELISA kit and may be used to determine metastasis
Immunohistochemistry method. development risk in the patients with
rectal cancer.
AC

OPN levels of 30 patient with ovarian OPN release was found higher in the
Ovarian endothelial cancer were measured patients with advanced stage of ovarian [73]
through PCR method. cancer when compared with early-stage
cancer patients.
An analysis was performed through OPN release level of the patients with
Colorectal biopsy from 64 patients with colorectal liver metastasis was detected higher [74]
cancer including 20 patients with than the patients without any
metastasis (liver) and 44 patients metastasis.
without any metastasis for evaluation
of OPN release according to metastasis
state of the patients with colorectal
cancer.
ACCEPTED MANUSCRIPT

3.6. Osteopontin and diabetes

It is reported that OPN is also active in formation of atherosclerotic plaque including diabetic

vascular complications [13, 75]. Osteopontin is highly secreted on medial layers of diabetic

rats and patient arteries [76]. Furthermore, as mentioned before, pro-inflammatory cytokines

which have a significant importance in development of diabetic complications increase in line

PT
with release of OPN [75]. Consequently, it is reported that OPN may play a key role in

development of insulin resistance by increasing macrophage accumulation in adipose tissue

RI
and promoting the inflammation formation [13]. Moreover, it is reported OPN level increases

SC
on vascular wall of diabetic individuals; and this may cause cell growth by increasing the
NU
OPN release [76]. It has also been reported that hyperglycemia stimulates OPN gene

expression in mesangial cells [77, 78]. However, it is not yet clear which mechanisms
MA

hyperglycemia enhances OPN gene expression, which is related to similar epigenetic

mechanisms described for NF- kB in blood vessels [79]. Hyperglycemia, a potent activator of
D

histone acetylation and methylation, has been reported to increase the expression of OPN
E

gene by increasing the activation of histone marks H3K9ac, H3K4me1 and H3K4me3 [80].
PT

In addition to these mechanisms, it is noted that OPN has many similar effects with GIP,
CE

including the regulation of adipocyte metabolism in fat tissue [13, 81] and the effect on

survival rates of β-cells in pancreatic islets [82, 83]. The incretin hormone GIP promotes
AC

pancreatic β-cell function by potentiating insulin secretion and β-cell proliferation [45]. GIP, a

potent stimulator of adipogenesis, is thought to cause insulin resistance by increasing OPN

release in the adipose tissue [84].

A study performed on the rats suggested that release of OPN was more in the rats with

diabetic nephropathy when compared with the control group [85]. Another study conducted
ACCEPTED MANUSCRIPT

on the rats concluded that the decrease in OPN level created an improvement in insulin

resistance [13]. In the study carried out by Daniele et al. (2014) on the patients with Type 2

diabetes as well as healthy individuals, it was detected that plasma OPN levels of the patients

with Type 2 diabetes were higher than the control group. Another outcome of such study was

the correlation between plasma OPN level and tumor necrosis factor-α (TNF-α) secretion

which mediate obesity-induced insulin resistance [86]. Nakamachi et al. found out in their

PT
study that proliferation-activated receptor (PPAR)α ligands suppressed expression of OPN in

RI
the individuals with Type 2 diabetes. Various studies report that OPN release increase in

SC
diabetes [87-89]. Gordin et al. (2014) carried out a study on the patients with Type 1 diabetes

and detected serum OPN is a strong predictor of incipient diabetic nephropathy. This study
NU
also suggests that serum OPN may be of clinical significance for the risk prediction of CVD

events in patients with Type 1 diabetes [90].


MA

In consideration of outcomes of the previous studies, it may be concluded that decreased


D

plasma levels of OPN concentrations may take an important role in prevention of diabetic
E

complications; however more comprehensive studies are needed [13, 75].


PT

3.7. Osteopontin and kidney stones


CE

Osteopontin is a protein which is synthesized from kidneys and released into the urine
AC

through epithelial cells and has important functions in formation of kidney stones [10, 91]. In

normal urine, magnesium tend to be crystallized through supersaturation in insufficient levels

of inhibitor molecules such as pyrophosphate and citrate [4]. Along with the effect of inhibitor

molecules on inhibition, they actually act through different proteins in the urine [4, 92]. Many

different studies report that OPN, as one of the aforesaid molecules, inhibit formation, growth

and accumulation of CaOx crystals [10, 93, 94]. In addition, OPN is reported to show an anti-
ACCEPTED MANUSCRIPT

lithogenic effect by inhibiting adherence of the crystals onto the renal epithelial cells [95].

However, there are different studies suggesting that OPN increases the risk of CaOx stone

formation by increasing renal tubular cell damage and crystal adherence onto the epithelium

[95-98].

Osteopontin is secreted from renal epithelial cells including loop of henle, distal tubular cells

PT
and papillary epithelial cells [4]. A negative correlation was found between urinary OPN level

and stone formation in different studies [24, 99]. Furthermore, kidney stones consists of

RI
proteins with a great amount. OPN is mentioned as one of the most important proteins within

SC
structure of calcium oxalate and calcium phosphate stones [100, 101]. Two basic mechanisms
NU
explain significantly lower OPN levels in the urine of the patients with kidney stone; the

hypothesis that lower levels of OPN which prevents formation, enlargement and accumulation
MA

of CaOx crystals or the hypthesis that OPN synthesis continues but less extraction in urine

because existing in the stone formation [10, 91, 92]. The second hypothesis is accepted more,
D

since it is known that expression of OPN increases in the patients whos kidney stones was
E

created experimentally and OPN strictly bind to hydroxyapetite [92, 102].


PT

There are studies focusing on genetic modifications on the OPN gene [103, 104]. Majority of
CE

these genetic studies address the polymorphisms on OPN gene [24, 104]. Liu et al. (2010)
AC

conducted a study on 249 patients with calcium stones and 247 healthy individuals and

detected -156 delG/G polymoprhism of the OPN gene significantly higher (p=0,037) in the

patient group. The aforesaid study concluded that such gene polymorphism level would be

used as a method to detect formation of calcium stones [24].

In a study carried out with 111 patients diagnosed with urinary system stone disease including

83 males and 28 females with an age range of 20 to 66 years, urinary OPN excretion of the
ACCEPTED MANUSCRIPT

patient group for 24 hours as well as the ratio to creatinine was found to be lower than the

control group (p<0.01) [99]. Another study evaluated urinary OPN excretion on 249 patients

with kidney stone as well as 247 healthy individuals. The study reported that urinary

excretion of OPN was significantly lower than the control group (p<0.001) [24]. On the other

hand, Tsuji et al. (2007) did not detect significant difference between urine OPN levels of the

participants in the study conducted on 120 patients with calcium oxalate stone and 40 healthy

PT
individuals (p>0,05) [105]. In line with such data, use of urinary OPN level may be

RI
considered as a biochemical parameter to diagnose the risk of stone formation. However,

SC
more human studies and reduction of measurement costs by new methods should be

performed for routine implementation.


NU
4. Conclusion
MA

OPN, which can be secreted by many tissues, has many controversial effects on health. It may

be considered that OPN increases the inflammation in acute and chronic inflammatory
E D

diseases due to its roles in increase of macrophage and T cells in inflammation sites.
PT

Furthermore, it may be concluded that OPN may reduce the risk of bone fractures by

modulating the adherence of osseous cells and matrix mineralization. Although the
CE

mechanisms are not clear yet, one of positive effects of OPN on health is anti-lithogenic
AC

effect. However, increased levels of OPN have some negative effects along with positive

effects on health. Some negative effects include that OPN stands as a risk factor for

cardiovascular diseases, diabetes and cancer. Along with the data presented, use of OPN level

may be considered as a new biochemical parameter to determine the risk of cardiovascular

diseases, diabetes and cancer. However, more human studies and reduction of measurement

costs by new methods should be performed for routine implementation.


ACCEPTED MANUSCRIPT

Conflict of interest

The authors have no relevant interests to declare.

Acknowledgment

Funding/support. No external funds supported this work.

PT
RI
SC
NU
MA
E D
PT
CE
AC
ACCEPTED MANUSCRIPT

References

1. Karpinsky G, Fatyga A, Krawczyk MA, Chamera M, Sande N, Szmyd D, Izycka-Swieszewska E,


Bien E: Osteopontin: its potential role in cancer of children and young adults. Biomarkers in
Medicine 2017, 11(4):389-402.
2. Clemente N, Raineri D, Cappellano G, Boggio E, Favero F, Soluri MF, Dianzani C, Comi C,
Dianzani U, Chiocchetti A: Osteopontin Bridging Innate and Adaptive Immunity in
Autoimmune Diseases. Journal of Immunology Research 2016, 2016.
3. Franzen A, Heinegård D: Isolation and characterization of two sialoproteins present only in
bone calcified matrix. Biochemical Journal 1985, 232(3):715-724.

PT
4. Mazzali M, Kipari T, Ophascharoensuk V, Wesson J, Johnson R, Hughes J: Osteopontin—a
molecule for all seasons. Qjm 2002, 95(1):3-13.
5. Chen Q, Shou P, Zhang L, Xu C, Zheng C, Han Y, Li W, Huang Y, Zhang X, Shao C: An

RI
Osteopontin‐Integrin Interaction Plays a Critical Role in Directing Adipogenesis and
Osteogenesis by Mesenchymal Stem Cells. Stem Cells 2014, 32(2):327-337.
6. Yoshitake H, Rittling SR, Denhardt DT, Noda M: Osteopontin-deficient mice are resistant to

SC
ovariectomy-induced bone resorption. Proceedings of the National Academy of Sciences
1999, 96(14):8156-8160.
7. Ashkar S, Weber GF, Panoutsakopoulou V, Sanchirico ME, Jansson M, Zawaideh S, Rittling SR,
NU
Denhardt DT, Glimcher MJ, Cantor H: Eta-1 (osteopontin): an early component of type-1
(cell-mediated) immunity. Science 2000, 287(5454):860-864.
8. Giachelli CM, Steitz S: Osteopontin: a versatile regulator of inflammation and
MA

biomineralization. Matrix Biology 2000, 19(7):615-622.


9. Berezin A, Kremzer A: Circulating osteopontin as a marker of early coronary vascular
calcification in type two diabetes mellitus patients with known asymptomatic coronary
artery disease. Atherosclerosis 2013, 229(2):475-481.
10. Wesson JA, Johnson RJ, Mazzali M, Beshensky AM, Stietz S, Giachelli C, Liaw L, Alpers CE,
D

Couser WG, Kleinman JG: Osteopontin is a critical inhibitor of calcium oxalate crystal
formation and retention in renal tubules. Journal of the American Society of Nephrology
E

2003, 14(1):139-147.
PT

11. Rangaswami H, Bulbule A, Kundu GC: Osteopontin: role in cell signaling and cancer
progression. Trends in cell biology 2006, 16(2):79-87.
12. Wai PY, Kuo PC: The role of osteopontin in tumor metastasis. Journal of Surgical Research
CE

2004, 121(2):228-241.
13. Nomiyama T, Perez-Tilve D, Ogawa D, Gizard F, Zhao Y, Heywood EB, Jones KL, Kawamori R,
Cassis LA, Tschöp MH: Osteopontin mediates obesity-induced adipose tissue macrophage
infiltration and insulin resistance in mice. The Journal of clinical investigation 2007,
AC

117(10):2877-2888.
14. Matsuzaki H, Katsumata S-i, Uehara M, Suzuki K, Miwa M: High-phosphorus diet induces
osteopontin expression of renal tubules in rats. Journal of clinical biochemistry and nutrition
2007, 41(3):179-183.
15. Wang X, Cheng M, Zhao M, Ge A, Guo F, Zhang M, Yang Y, Liu L, Yang N: Differential effects
of high-fat-diet rich in lard oil or soybean oil on osteopontin expression and inflammation
of adipose tissue in diet-induced obese rats. European journal of nutrition 2013, 52(3):1181-
1189.
16. Singh A, Pezeshki A, Zapata RC, Yee NJ, Knight CG, Tuor UI, Chelikani PK: Diets enriched in
whey or casein improve energy balance and prevent morbidity and renal damage in salt-
loaded and high-fat-fed spontaneously hypertensive stroke-prone rats. The Journal of
nutritional biochemistry 2016, 37:47-59.
ACCEPTED MANUSCRIPT

17. Lund SA, Giachelli CM, Scatena M: The role of osteopontin in inflammatory processes.
Journal of cell communication and signaling 2009, 3(3-4):311-322.
18. Standal T, Borset M, Sundan A: Role of osteopontin in adhesion, migration, cell survival and
bone remodeling. Exp Oncol 2004, 26(3):179-184.
19. Denhardt DT, Guo X: Osteopontin: a protein with diverse functions. The FASEB journal 1993,
7(15):1475-1482.
20. Denhardt DT, Noda M: Osteopontin expression and function: role in bone remodeling.
Journal of Cellular Biochemistry 1998, 72(S30‒31):92-102.
21. Chackalaparampil I, Peri A, Nemir M, Mckee MD, Lin P-H, Mukherjee BB, Mukherjee AB: Cells
in vivo and in vitro from osteopetrotic mice homozygous for c-src disruption show
suppression of synthesis of osteopontin, a multifunctional extracellular matrix protein.

PT
Oncogene 1996, 12(7):1457-1467.
22. Coppola D, Szabo M, Boulware D, Muraca P, Alsarraj M, Chambers AF, Yeatman TJ:
Correlation of osteopontin protein expression and pathological stage across a wide variety
of tumor histologies. Clinical cancer research 2004, 10(1):184-190.

RI
23. Ophascharoensuk V, Giachelli CM, Gordon K, Hughes J, Pichler R, Brown P, Liaw L, Schmidt R,
Shankland SJ, Alpers CE: Obstructive uropathy in the mouse: role of osteopontin in

SC
interstitial fibrosis and apoptosis. Kidney international 1999, 56(2):571-580.
24. Liu C-C, Huang S-P, Tsai L-Y, Wu W-J, Juo S-HH, Chou Y-H, Huang C-H, Wu M-T: The impact of
osteopontin promoter polymorphisms on the risk of calcium urolithiasis. Clinica chimica
NU
acta 2010, 411(9):739-743.
25. Şentürk N: Kütanöz inflamasyon. Archives of the Turkish Dermatology &
Venerology/Turkderm 2013.
26. Nau GJ, Liaw L, Chupp GL, Berman JS, Hogan BL, Young RA: Attenuated Host Resistance
MA

againstMycobacterium bovis BCG Infection in Mice Lacking Osteopontin. Infection and


immunity 1999, 67(8):4223-4230.
27. Gómez-Ambrosi J, Catalán V, Ramírez B, Rodríguez A, Colina I, Silva C, Rotellar F, Mugueta C,
Gil MaJ, Cienfuegos JA: Plasma osteopontin levels and expression in adipose tissue are
D

increased in obesity. The Journal of Clinical Endocrinology & Metabolism 2007, 92(9):3719-
3727.
E

28. Kiefer FW, Zeyda M, Todoric J, Huber J, Geyeregger R, Weichhart T, Aszmann O, Ludvik B,
Silberhumer GR, Prager G: Osteopontin expression in human and murine obesity: extensive
PT

local up-regulation in adipose tissue but minimal systemic alterations. Endocrinology 2008,
149(3):1350-1357.
29. Schaller G, Aso Y, Schernthaner G-H, Kopp H-P, Inukai T, Kriwanek S, Schernthaner G:
CE

Increase of osteopontin plasma concentrations after bariatric surgery independent from


inflammation and insulin resistance. Obesity surgery 2009, 19(3):351.
30. Nau GJ, Guilfoile P, Chupp GL, Berman JS, Kim SJ, Kornfeld H, Young RA: A chemoattractant
AC

cytokine associated with granulomas in tuberculosis and silicosis. Proceedings of the


National Academy of Sciences 1997, 94(12):6414-6419.
31. Nau GJ, Chupp GL, Emile J-F, Jouanguy E, Berman JS, Casanova J-L, Young RA: Osteopontin
expression correlates with clinical outcome in patients with mycobacterial infection. The
American journal of pathology 2000, 157(1):37-42.
32. 泉啓介: Baseline serum osteopontin levels predict the clinical effectiveness of tocilizumab
but not infliximab in biologic-naïve patients with rheumatoid arthritis: a single-center
prospective study at 1 year: the Keio First-Bio Cohort Study. 慶應義塾大学; 2016.
33. Iida T, Wagatsuma K, Hirayama D, Nakase H: Is Osteopontin a Friend or Foe of Cell Apoptosis
in Inflammatory Gastrointestinal and Liver Diseases? International journal of molecular
sciences 2017, 19(1):7.
34. Lenton S, Seydel T, Nylander T, Holt C, Härtlein M, Teixeira S, Zaccai G: Dynamic footprint of
sequestration in the molecular fluctuations of osteopontin. Journal of The Royal Society
Interface 2015, 12(110):20150506.
ACCEPTED MANUSCRIPT

35. Reinholt FP, Hultenby K, Oldberg A, Heinegård D: Osteopontin--a possible anchor of


osteoclasts to bone. Proceedings of the National Academy of Sciences 1990, 87(12):4473-
4475.
36. Ross FP, Chappel J, Alvarez J, Sander D, Butler W, Farach-Carson M, Mintz K, Robey PG,
Teitelbaum S, Cheresh D: Interactions between the bone matrix proteins osteopontin and
bone sialoprotein and the osteoclast integrin alpha v beta 3 potentiate bone resorption.
Journal of Biological Chemistry 1993, 268(13):9901-9907.
37. McKee M, Nanci A: Osteopontin at mineralized tissue interfaces in bone, teeth, and
osseointegrated implants: ultrastructural distribution and implications for mineralized
tissue formation, turnover, and repair. Microscopy research and technique 1996, 33(2):141-
164.

PT
38. Liaw L, Birk DE, Ballas CB, Whitsitt JS, Davidson JM, Hogan BL: Altered wound healing in mice
lacking a functional osteopontin gene (spp1). Journal of Clinical Investigation 1998,
101(7):1468.
39. Rittling SR, Matsumoto HN, Mckee MD, Nanci A, An XR, Novick KE, Kowalski AJ, Noda M,

RI
Denhardt DT: Mice lacking osteopontin show normal development and bone structure but
display altered osteoclast formation in vitro. Journal of Bone and Mineral Research 1998,

SC
13(7):1101-1111.
40. Chellaiah MA, Soga N, Swanson S, McAllister S, Alvarez U, Wang D, Dowdy SF, Hruska KA:
Rho-A is critical for osteoclast podosome organization, motility, and bone resorption.
NU
Journal of Biological Chemistry 2000, 275(16):11993-12002.
41. Giachelli CM, Bae N, Almeida M, Denhardt DT, Alpers CE, Schwartz SM: Osteopontin is
elevated during neointima formation in rat arteries and is a novel component of human
atherosclerotic plaques. Journal of Clinical Investigation 1993, 92(4):1686.
MA

42. Steitz SA, Speer MY, Curinga G, Yang H-Y, Haynes P, Aebersold R, Schinke T, Karsenty G,
Giachelli CM: Smooth muscle cell phenotypic transition associated with calcification.
Circulation research 2001, 89(12):1147-1154.
43. Leali D, Dell’Era P, Stabile H, Sennino B, Chambers AF, Naldini A, Sozzani S, Nico B, Ribatti D,
D

Presta M: Osteopontin (Eta-1) and fibroblast growth factor-2 cross-talk in angiogenesis. The
Journal of Immunology 2003, 171(2):1085-1093.
E

44. McIntosh CH, Widenmaier S, Kim S-J: Pleiotropic actions of the incretin hormones. In:
Vitamins & Hormones. Volume 84, edn.: Elsevier; 2010: 21-79.
PT

45. Lyssenko V, Eliasson L, Kotova O, Pilgaard K, Wierup N, Salehi A, Wendt A, Jonsson A, De


Marinis YZ, Berglund LM: Pleiotropic effects of GIP on islet function involve osteopontin.
Diabetes 2011, 60(9):2424-2433.
CE

46. Nitz I, Fisher E, Weikert C, Burwinkel B, Li Y, Möhlig M, Boeing H, Schreiber S, Schrezenmeir J,


Döring F: Association analyses of GIP and GIPR polymorphisms with traits of the metabolic
syndrome. Molecular nutrition & food research 2007, 51(8):1046-1052.
AC

47. Berglund LM, Lyssenko V, Ladenvall C, Kotova O, Edsfeldt A, Pilgaard K, Alkayyali S, Brøns C,
Forsblom C, Jonsson A: Glucose-dependent insulinotropic polypeptide (GIP) stimulates
osteopontin expression in the vasculature via endothelin-1 and CREB. Diabetes
2015:db150122.
48. Omar B, Banke E, Guirguis E, Åkesson L, Manganiello V, Lyssenko V, Groop L, Gomez MF,
Degerman E: Regulation of the pro-inflammatory cytokine osteopontin by GIP in
adipocytes–a role for the transcription factor NFAT and phosphodiesterase 3B. Biochemical
and biophysical research communications 2012, 425(4):812-817.
49. Beadenkopf W: Calcification in the coronary arteries and its relationship to arteriosclerosis
and myocardial infarction. Am J Roentgenol Radium Ther Nucl Med 1964, 92:865-871.
50. Fitzpatrick L, Severson A, Edwards W, Ingram R: Diffuse calcification in human coronary
arteries. Association of osteopontin with atherosclerosis. Journal of Clinical Investigation
1994, 94(4):1597.
ACCEPTED MANUSCRIPT

51. Abdalrhim AD, Marroush TS, Austin EE, Gersh BJ, Solak N, Rizvi SA, Bailey KR, Kullo IJ: Plasma
Osteopontin Levels and Adverse Cardiovascular Outcomes in the PEACE Trial. PloS one
2016, 11(6):e0156965.
52. Dimitrow PP, Jawien M, Gackowski A: The influence of statins on levels of calcification
biomarkers in patients with aortic sclerosis or mild aortic stenosis. J Heart Valve Dis 2011,
20(1).
53. Srivatsa S, Harrity P, Maercklein P, Kleppe L, Veinot J, Edwards W, Johnson C, Fitzpatrick L:
Increased cellular expression of matrix proteins that regulate mineralization is associated
with calcification of native human and porcine xenograft bioprosthetic heart valves.
Journal of Clinical Investigation 1997, 99(5):996.
54. Pritzker L, Scatena M, Giachelli C: The role of osteoprotegerin and tumor necrosis factor-

PT
related apoptosis-inducing ligand in human microvascular endothelial cell survival.
Molecular biology of the cell 2004, 15(6):2834-2841.
55. Kurban S, Mehmetoglu I: Osteoprotegerin, RANK and RANK ligand. TURKISH JOURNAL OF
BIOCHEMISTRY-TURK BIYOKIMYA DERGISI 2007, 32(4):178-184.

RI
56. McKee MD, Pedraza CE, Kaartinen MT: Osteopontin and wound healing in bone. Cells
Tissues Organs 2011, 194(2-4):313-319.

SC
57. Lin Y-H, Huang C-J, Chao J-R, Chen S-T, Lee S-F, Yen JJ-Y, Yang-Yen H-F: Coupling of
osteopontin and its cell surface receptor CD44 to the cell survival response elicited by
interleukin-3 or granulocyte-macrophage colony-stimulating factor. Molecular and cellular
NU
biology 2000, 20(8):2734-2742.
58. Lenga Y, Koh A, Perera AS, McCulloch CA, Sodek J, Zohar R: Osteopontin expression is
required for myofibroblast differentiation. Circulation research 2008, 102(3):319-327.
59. Brown LF, Papadopoulos-Sergiou A, Berse B, Manseau EJ, Tognazzi K, Perruzzi CA, Dvorak HF,
MA

Senger DR: Osteopontin expression and distribution in human carcinomas. The American
journal of pathology 1994, 145(3):610.
60. Guo X, Zhang YP, Mitchell DA, Denhardt DT, Chambers AF: Identification of a ras-activated
enhancer in the mouse osteopontin promoter and its interaction with a putative ETS-
D

related transcription factor whose activity correlates with the metastatic potential of the
cell. Molecular and cellular biology 1995, 15(1):476-487.
E

61. Correa P, Haenszel W, Cuello C, Zavala D, Fontham E, Zarama G, Tannenbaum S, Collazos T,


Ruiz B: Gastric precancerous process in a high risk population: cohort follow-up. Cancer
PT

Research 1990, 50(15):4737-4740.


62. Chang WL, Yang HB, Cheng HC, Chuang CH, Lu PJ, Sheu BS: Increased gastric osteopontin
expression by Helicobacter pylori Infection can correlate with more severe gastric
CE

inflammation and intestinal metaplasia. Helicobacter 2011, 16(3):217-224.


63. Flamant S, Kortulewski T, Dugray A, Bonnet M-L, Guillier M, Guilhot F, Bourhis J-H,
Vainchenker W, Tronik-Le Roux D, Turhan A: Osteopontin is upregulated by BCR-ABL.
AC

Biochemical and biophysical research communications 2005, 333(4):1378-1384.


64. Lee J-L, Wang M-J, Sudhir P-R, Chen G-D, Chi C-W, Chen J-Y: Osteopontin promotes integrin
activation through outside-in and inside-out mechanisms: OPN-CD44V interaction
enhances survival in gastrointestinal cancer cells. Cancer research 2007, 67(5):2089-2097.
65. Lee S-H, Park J-W, Go D-M, Kim HK, Kwon H-J, Han S-U, Kim D-Y: Ablation of osteopontin
suppresses N-methyl-N-nitrosourea and Helicobacter pylori-induced gastric cancer
development in mice. Carcinogenesis 2015, 36(12):1550-1560.
66. Chagan-Yasutan H, Tsukasaki K, Takahashi Y, Oguma S, Harigae H, Ishii N, Zhang J, Fukumoto
M, Hattori T: Involvement of osteopontin and its signaling molecule CD44 in
clinicopathological features of adult T cell leukemia. Leukemia research 2011, 35(11):1484-
1490.
67. Liersch R, Gerss J, Schliemann C, Bayer M, Schwöppe C, Biermann C, Appelmann I, Kessler T,
Löwenberg B, Büchner T: Osteopontin is a prognostic factor for survival of acute myeloid
leukemia patients. Blood 2012, 119(22):5215-5220.
ACCEPTED MANUSCRIPT

68. Psyrri A, Kalogeras KT, Wirtz RM, Kouvatseas G, Karayannopoulou G, Goussia A, Zagouri F,
Veltrup E, Timotheadou E, Gogas H: Association of osteopontin with specific prognostic
factors and survival in adjuvant breast cancer trials of the Hellenic Cooperative Oncology
Group. Journal of translational medicine 2017, 15(1):30.
69. Di Bartolomeo M, Pietrantonio F, Pellegrinelli A, Martinetti A, Mariani L, Daidone MG, Bajetta
E, Pelosi G, de Braud F, Floriani I: Osteopontin, E-cadherin, and β-catenin expression as
prognostic biomarkers in patients with radically resected gastric cancer. Gastric cancer
2016, 19(2):412-420.
70. Tilli T, Thuler L, Matos A, Coutinho-Camillo C, Soares F, da Silva E, Neves A, Goulart L, Gimba
E: Expression analysis of osteopontin mRNA splice variants in prostate cancer and benign
prostatic hyperplasia. Experimental and molecular pathology 2012, 92(1):13-19.

PT
71. Mack PC, Redman MW, Chansky K, Williamson SK, Farneth NC, Lara Jr PN, Franklin WA, Le Q-
T, Crowley JJ, Gandara DR: Lower osteopontin plasma levels are associated with superior
outcomes in advanced non–small-cell lung cancer patients receiving platinum-based
chemotherapy: SWOG Study S0003. Journal of Clinical Oncology 2008, 26(29):4771-4776.

RI
72. Debucquoy A, Goethals L, Geboes K, Roels S, Mc Bride WH, Haustermans K: Molecular
responses of rectal cancer to preoperative chemoradiation. Radiotherapy and oncology

SC
2006, 80(2):172-177.
73. Hashiguchi Y, Tsuda H, Bandera CA, Nishimura S, Inoue T, Kawamura N, Berkowitz RS, Mok
SC: Comparison of osteopontin expression in endometrioid endometrial cancer and ovarian
NU
endometrioid cancer. Medical Oncology 2006, 23(2):205-212.
74. Huang J, Pan C, Hu H, Zheng S, Ding L: Osteopontin-enhanced hepatic metastasis of
colorectal cancer cells. PloS one 2012, 7(10):e47901.
75. Yan X, Sano M, Lu L, Wang W, Zhang Q, Zhang R, Wang L, Chen Q, Fukuda K, Shen W: Plasma
MA

concentrations of osteopontin, but not thrombin-cleaved osteopontin, are associated with


the presence and severity of nephropathy and coronary artery disease in patients with
type 2 diabetes mellitus. Cardiovascular diabetology 2010, 9(1):70.
76. Takemoto M, Yokote K, Nishimura M, Shigematsu T, Hasegawa T, Kon S, Uede T, Matsumoto
D

T, Saito Y, Mori S: Enhanced expression of osteopontin in human diabetic artery and


analysis of its functional role in accelerated atherogenesis. Arteriosclerosis, Thrombosis, and
E

Vascular Biology 2000, 20(3):624-628.


77. Sodhi CP, Phadke SA, Batlle D, Sahai A: Hypoxia and high glucose cause exaggerated
PT

mesangial cell growth and collagen synthesis: role of osteopontin. American Journal of
Physiology-Renal Physiology 2001, 280(4):F667-F674.
78. Sun J, Xu Y, Deng H, Sun S, Dai Z, Sun Y: Involvement of osteopontin upregulation on
CE

mesangial cells growth and collagen synthesis induced by intermittent high glucose. Journal
of cellular biochemistry 2010, 109(6):1210-1221.
79. El-Osta A, Brasacchio D, Yao D, Pocai A, Jones PL, Roeder RG, Cooper ME, Brownlee M:
AC

Transient high glucose causes persistent epigenetic changes and altered gene expression
during subsequent normoglycemia. Journal of Experimental Medicine 2008, 205(10):2409-
2417.
80. Cai M, Bompada P, Atac D, Laakso M, Groop L, De Marinis Y: Epigenetic regulation of
glucose-stimulated osteopontin (OPN) expression in diabetic kidney. Biochemical and
biophysical research communications 2016, 469(1):108-113.
81. Kim S-J, Nian C, McIntosh CH: GIP increases human adipocyte LPL expression through CREB
and TORC2-mediated trans-activation of the LPL gene. Journal of lipid research 2010,
51(11):3145-3157.
82. Arafat HA, Katakam AK, Chipitsyna G, Gong Q, Vancha AR, Gabbeta J, Dafoe DC: Osteopontin
protects the islets and β-cells from interleukin-1 β-mediated cytotoxicity through negative
feedback regulation of nitric oxide. Endocrinology 2007, 148(2):575-584.
83. Kim S-J, Winter K, Nian C, Tsuneoka M, Koda Y, McIntosh CH: Glucose-dependent
insulinotropic polypeptide (GIP) stimulation of pancreatic β-cell survival is dependent upon
ACCEPTED MANUSCRIPT

phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PKB) signaling, inactivation of the


forkhead transcription factor Foxo1, and down-regulation of bax expression. Journal of
Biological Chemistry 2005, 280(23):22297-22307.
84. Ahlqvist E, Osmark P, Kuulasmaa T, Pilgaard K, Omar B, Brøns C, Kotova O, Zetterqvist AV,
Stančáková A, Jonsson A: Link between GIP and osteopontin in adipose tissue and insulin
resistance. Diabetes 2013, 62(6):2088-2094.
85. Fischer JW, Tschöpe C, Reinecke A, Giachelli CM, Unger T: Upregulation of osteopontin
expression in renal cortex of streptozotocin-induced diabetic rats is mediated by
bradykinin. Diabetes 1998, 47(9):1512-1518.
86. Daniele G, Mendoza RG, Winnier D, Fiorentino T, Pengou Z, Cornell J, Andreozzi F, Jenkinson
C, Cersosimo E, Federici M: The inflammatory status score including IL-6, TNF-α,

PT
osteopontin, fractalkine, MCP-1 and adiponectin underlies whole-body insulin resistance
and hyperglycemia in type 2 diabetes mellitus. Acta diabetologica 2014, 51(1):123-131.
87. Kelly DJ, Wilkinson‐Berka JL, Ricardo SD, Cox AJ, Gilbert RE: Progression of tubulointerstitial
injury by osteopontin‐induced macrophage recruitment in advanced diabetic nephropathy

RI
of transgenic (mRen‐2) 27 rats. Nephrology Dialysis Transplantation 2002, 17(6):985-991.
88. Kiefer FW, Zeyda M, Gollinger K, Pfau B, Neuhofer A, Weichhart T, Säemann MD, Geyeregger

SC
R, Schlederer M, Kenner L: Neutralization of osteopontin inhibits obesity-induced
inflammation and insulin resistance. Diabetes 2010, 59(4):935-946.
89. Takemoto M, Yokote K, Yamazaki M, Ridall AL, Butler WT, Matsumoto T, Tamura K, Saito Y,
NU
Mori S: Enhanced expression of osteopontin by high glucose in cultured rat aortic smooth
muscle cells. Biochemical and biophysical research communications 1999, 258(3):722-726.
90. Gordin D, Forsblom C, Panduru NM, Thomas MC, Bjerre M, Soro-Paavonen A, Tolonen N,
Sandholm N, Flyvbjerg A, Harjutsalo V: Osteopontin is a strong predictor of incipient
MA

diabetic nephropathy, cardiovascular disease, and all-cause mortality in patients with type
1 diabetes. Diabetes Care 2014, 37(9):2593-2600.
91. Hudkins KL, Giachelli CM, Cui Y, Couser WG, Johnson RJ, Alpers CE: Osteopontin expression
in fetal and mature human kidney. Journal of the American Society of Nephrology 1999,
D

10(3):444-457.
92. Yasui T, Fujita K, Hayashi Y, Ueda K, Kon S, Maeda M, Uede T, Kohri K: Quantification of
E

osteopontin in the urine of healthy and stone-forming men. Urological research 1999,
27(4):225-230.
PT

93. Asplin JR, Arsenault D, Parks JH, Coe FL, Hoyer JR: Contribution of human uropontin to
inhibition of calcium oxalate crystallization. Kidney international 1998, 53(1):194-199.
94. Kleinman JG, Beshensky A, Worcester EM, Brown D: Expression of osteopontin, a urinary
CE

inhibitor of stone mineral crystal growth, in rat kidney. Kidney international 1995,
47(6):1585-1596.
95. Okada A, Nomura S, Saeki Y, Higashibata Y, Hamamoto S, Hirose M, Itoh Y, Yasui T, Tozawa K,
AC

Kohri K: Morphological conversion of calcium oxalate crystals into stones is regulated by


osteopontin in mouse kidney. Journal of bone and mineral research 2008, 23(10):1629-1637.
96. Hirose M, Tozawa K, Okada A, Hamamoto S, Higashibata Y, Gao B, Hayashi Y, Shimizu H,
Kubota Y, Yasui T: Role of osteopontin in early phase of renal crystal formation:
immunohistochemical and microstructural comparisons with osteopontin knock-out mice.
Urological research 2012, 40(2):121-129.
97. Hamamoto S, Yasui T, Okada A, Hirose M, Matsui Y, Kon S, Sakai F, Kojima Y, Hayashi Y,
Tozawa K: Crucial role of the cryptic epitope SLAYGLR within osteopontin in renal crystal
formation of mice. Journal of Bone and Mineral Research 2011, 26(12):2967-2977.
98. Asselman M, Verhulst A, de Broe ME, Verkoelen CF: Calcium oxalate crystal adherence to
hyaluronan-, osteopontin-, and CD44-expressing injured/regenerating tubular epithelial
cells in rat kidneys. Journal of the American Society of Nephrology 2003, 14(12):3155-3166.
99. Yaman F, Aybek H, Aybek Z: Üriner sistem taş hastalığı etiyopatogenezinde fetuin-a ve
osteopotin. Pamukkale Üniversitesi; 2011.
ACCEPTED MANUSCRIPT

100. Tawada T, Fujita K, Sakakura T, Shibutani T, Nagata T, Iguchi M, Kohri K: Distribution of


osteopontin and calprotectin as matrix protein in calcium-containing stone. Urological
research 1999, 27(4):238-242.
101. Yasui T, Fujita K, Sasaki S, Sato M, Sugimoto M, Hirota S, Kitamura Y, Nomura S, Kohri K:
Expression of bone matrix proteins in urolithiasis model rats. Urological research 1999,
27(4):255-261.
102. Kleinman JG, Wesson JA, Hughes J: Osteopontin and calcium stone formation. Nephron
Physiology 2004, 98(2):p43-p47.
103. Yamate T, Tsuji H, Amasaki N, Iguchi M, Kurita T, Kohri K: Analysis of osteopontin DNA in
patients with urolithiasis. Urological Research 2000, 28(3):159-166.
104. Gao B, Yasui T, Itoh Y, Li Z, Okada A, Tozawa K, Hayashi Y, Kohri K: Association of osteopontin

PT
gene haplotypes with nephrolithiasis. Kidney international 2007, 72(5):592-598.
105. Tsuji H, Tohru U, Hirotsugu U, Masanori I, Yuji H, Takashi K: Urinary concentration of
osteopontin and association with urinary supersaturation and crystal formation.
International journal of urology 2007, 14(7):630-634.

RI
SC
NU
MA
E D
PT
CE
AC
ACCEPTED MANUSCRIPT

Highlights

 Osteopontin is a significant chemical attractant for macrophages and T cells.

 Osteopontin reduce the risk of bone fractures through the different mechanisms.

 Osteopontin increases risk of atherosclerotic plaque formation leading to calcification

in arteries.

PT
 Plasma osteopontin levels correlate with tumor necrosis factor-α (TNF-α) secretion.

 Osteopontin plays important roles in wound healing due to its effects on cell viability.

RI
 Use of urinary osteopontin level may be considered as a biochemical parameter to

SC
diagnose the risk of stone formation. NU
MA
E D
PT
CE
AC
Graphics Abstract
Figure 1
Figure 2
Figure 3

You might also like