Next Article in Journal
Long Non-Coding RNAs Target Pathogenetically Relevant Genes and Pathways in Rheumatoid Arthritis
Next Article in Special Issue
Osteopontin is An Important Regulative Component of the Fetal Bone Marrow Hematopoietic Stem Cell Niche
Previous Article in Journal
Interplay between Phosphatases and the Anaphase-Promoting Complex/Cyclosome in Mitosis
Previous Article in Special Issue
Osteopontin in the Cerebrospinal Fluid of Patients with Severe Aneurysmal Subarachnoid Hemorrhage
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Osteopontin as a Link between Inflammation and Cancer: The Thorax in the Spotlight

by
Anne-Sophie Lamort
1,*,†,
Ioanna Giopanou
2,†,
Ioannis Psallidas
3 and
Georgios T. Stathopoulos
1,2,*
1
Comprehensive Pneumology Center and Institute for Lung Biology and Disease, University Hospital, Ludwig-Maximilians University of Munich and Helmholtz Center Munich, Member of the German Center for Lung Research, Max-Lebsche-Platz 31, 81377 Munich, Bavaria, Germany
2
Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Biomedical Sciences Research Center, 1 Asklepiou Str., University Campus, 26504 Rio, Achaia, Greece
3
Lungs for Living Research Centre, UCL Respiratory, University College London, London WC1E6BT, UK
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2019, 8(8), 815; https://doi.org/10.3390/cells8080815
Submission received: 14 June 2019 / Revised: 25 July 2019 / Accepted: 31 July 2019 / Published: 2 August 2019
(This article belongs to the Special Issue Adding New Pieces in the Osteopontin Puzzle)

Abstract

:
The glycoprotein osteopontin (OPN) possesses multiple functions in health and disease. To this end, osteopontin has beneficial roles in wound healing, bone homeostasis, and extracellular matrix (ECM) function. On the contrary, osteopontin can be deleterious for the human body during disease. Indeed, osteopontin is a cardinal mediator of tumor-associated inflammation and facilitates metastasis. The purpose of this review is to highlight the importance of osteopontin in malignant processes, focusing on lung and pleural tumors as examples.

1. Introduction

Osteopontin (OPN), also called secreted phosphoprotein 1 (SPP1), is encoded by the human gene SPP1 and its murine counterpart Spp1 [1,2]. Alternative splicing of the human OPN transcript is producing three isoforms called OPN-a, OPN-b, and OPN-c [3] that can be secretory or intracellular [4,5]. OPN is a glycoprotein and presents different post-translational modifications according to the tissue of expression [6,7]. The biological functions of OPN are diverse and specific to a spectrum of physiological and disease conditions. In physiological processes such as wound healing, secreted OPN is involved in cell adhesion to the extracellular matrix via its integrin-binding sequence [8,9], whereas intracellular OPN is involved in mitosis [5]. During chronic inflammation associated with infection and cancer development, OPN regulates host immunity [10] and is a poor prognosticator of survival by enhancing the proliferation and by preventing apoptosis of tumor cells [11]. In non-small cell lung cancers (NSCLCs), OPN induces VEGF expression and facilitates disease progression [12]. Altogether, the multifaceted functions of OPN render it a potential biomarker and therapeutic target, as well as a molecule for which new research tools such as neutralizing antibodies and small interfering RNAs are constantly developed [13,14]. The first part of this review focuses on the OPN gene, the protein isoforms, and their tissue distribution. Subsequently, OPN functions in health and disease are summarized with an emphasis on inflammatory and malignant processes of the chest. Finally, the diverse tools and strategies developed to study OPN biology are discussed.

2. Materials and Methods

The knowledge base of this review is based on PubMed searches performed for the present manuscript (https://www.ncbi.nlm.nih.gov/pubmed/; accession date: 14.11.2018). The first search was done using the terms “(osteopontin OR SPP1 OR OPN OR (secreted phosphoprotein 1)) AND cancer” and the limit of publication date within the last five years, yielding 876 results, of which the top 50 sorted by relevance and the top 50 sorted by date were manually reviewed. Their titles and abstracts were studied, and 29 publications were selected for full-text review. Another search employed the terms “(osteopontin OR SPP1 OR OPN OR (secreted phosphoprotein 1)) AND (lung cancer)” and the limit of publication date within the last five years, yielding 128 results, of which the top 50 sorted by relevance and the top 50 sorted by date that was non-redundant with the first search were manually analyzed by screening their titles and abstracts, and 38 papers were selected for full-text review. A final search using the terms “(osteopontin OR SPP1 OR OPN OR (secreted phosphoprotein 1)) AND inflammation” and the limit of publication date within the last five years yielded 514 results, of which the top 50 sorted by relevance and the top 50 sorted by date were manually reviewed. Their titles and abstracts were also screened, and 38 publications were selected for full-text review. Additional searches were performed in the U.S. Clinical Trial Database (https://clinicaltrials.gov/; date of accession: 14.11.2018) using the same terms, providing 25 registered trials, all of which were fully reviewed.

3. Review

3.1. Physiology of Osteopontin in Humans and Experimental Animals

3.1.1. Osteopontin Genes, Transcripts, and Proteins

OPN was first identified in malignant cells in 1979 [15]. In normal tissues, OPN was described in 1985 as bone sialoprotein 1 only to be renamed to OPN the following year [16,17]. OPN, also called nephropontin and SPP1, is encoded by the human gene SPP1 [1]. The SPP1 gene is located on chromosome 4 in locus 4q13.22 (Figure 1) that is encoding proteins of the small integrin-binding ligand N-linked glycoprotein (SIBLING) family [18,19]. The human SPP1 gene comprises seven exons that are alternatively spliced producing different variants [1,20]. The alternative splicing of OPN was described in 2006 with three alternatives forms of the protein [21]. OPN-a is considered the canonical transcript, OPN-b variant is lacking exon 5, and OPN-c variant is missing exon 4; translation of these variants leads to the expression of three different proteins called OPN-a (314 aa), OPN-b (300 aa), and OPN-c (287 aa) [3]. These proteins are negatively charged due to the presence of multiple aspartate residues and their secondary structures are undefined [22]. The murine Spp1 orthologue is located on chromosome 5 in locus qE5 [2]. The eight exons which compose the gene correspond to a protein composed of 294 amino acids [23].

3.1.2. Tissue-Restricted Patterns of Osteopontin Expression and Processing

The OPN protein is produced in a wide range of tissues all over the body in physiologic and disease conditions (Figure 2). However, only a few cell types can express OPN, such as osteoblasts of osseous tissue [24,25,26,27], fibroblasts and activated macrophages of healing wounds [9,28,29], and dendritic, lymphoid, and mononuclear cells of the immune system [30,31,32,33]. Secretory OPN (sOPN) is present in biologic fluids like human serum (average level of 159 ng/mL) [34], bovine milk (average levels 18-138 mg/L) [35], and human urine (average level of 4 µg/mL) [36]. However, intracellular OPN (iOPN) is also expressed [5,37,38] from an alternative downstream transcription initiation site that results in skipping of the first 16 amino acids that function as a membrane localization signal peptide [5,39]. In the lungs, OPN is observed in the sputum (average level of 53 pg/mL) [40] and bronchoalveolar lavage fluid (BALF; average level of 1.1 ng/mL) [41] of healthy individuals, constitutively secreted by airway epithelial cells and alveolar macrophages and exerting diverse functions dependent on its phosphorylation status [42]. In general, sOPN presents different and specific patterns of post-translational modifications according to its cell of origin. For instance, human milk OPN exhibits 36 phosphoserines, 2 phosphothreonines, and 5 O-glycosylated threonines [6], whereas HEK293T cell-derived OPN displays 26 phosphothreonines and 7 O-glycosylated threonines [7]. Unfortunately, there is a lack of information regarding the post-translational modification status of OPN in the lungs. Little information is available and concerns five O-glycosylations, as well as their location and their function [43]. Post-translational OPN modifications result in different functions, but despite the growing literature on the topic, OPN structure–function relationships remain incompletely understood.

3.1.3. Regulation of Osteopontin Expression

OPN expression is regulated by various transcription factors (Table 1) which can link regulatory sequences such as AML-1 and C/EBPα that bind to the E-box motif in the region −170 to −127 of the SPP1 promoter [44]. Other potential regulatory sequences in the SPP1 promoter have been identified, including a TATA-like sequence (nucleotides −27 to −22), vitamin-D-responsive (VDR)-like motifs (nucleotides −1892 to −1878 and −698 to −684), an AP-1 binding sequence (TGACACA, nucleotides −78 to −72), and an Ets-1 motif (nucleotides −47 to −39) [45]. Diverse stimuli regulate OPN expression including mechanical stress, hormones, inflammatory mediators, and others listed in Table 1. For instance, in alveolar macrophages, the E3 ligase von Hippel–Lindau protein (VHL) promotes OPN expression by epigenetic modifications [46]. On the contrary, in lung adenocarcinoma-derived A549 cells, the transcription factor Smad4 can bind the OPN gene promoter and inhibit its transcription [47].
In the mouse, similar regulators exist, such as vitamin D3 that can downregulate OPN [58] or transcription factors that can activate OPN expression via promoter binding [59].

3.1.4. Osteopontin as a Matricellular Component

OPN is a significant component of the extracellular matrix (ECM) that regulates matrix interactions and cell adhesion. OPN interacts with ECM elements like collagen and fibronectin, as well as with calcium due to its Ca2+ binding sequence [60]. For cell adhesion, OPN possesses integrin-binding sites—an RGD sequence in position 159–161 recognized by integrins αvβ1, αvβ3, αvβ5, α5β1, and α8β1, and the SVVYGLR motif (amino acids 162–168) bound by integrins α9β1, α4β1, and α4β7 [8]. Moreover, the C-terminus of OPN is recognized by the CD44 splice variants CD44v3, CD44v6, and CD44v7 [8]. OPN cleavage by matrix metalloprotease 3 (MMP3), MMP7, and thrombin renders the above motifs more accessible and enhances binding efficiency [4,61]. In lung cancer, the absence of integrin β3 allows OPN-a to stimulate cell growth via the CD44/NFκB pathway, and at high levels of integrin β3 OPN-a inhibits lung cancer cell growth [62].

3.1.5. Osteopontin as a Secreted Protein

OPN secreted by immune cells promotes angiogenesis by binding integrin αvβ3 and activating the PI3K/AKT and ERK pathways on endothelial cells, thereby stimulating VEGF production [63]. Myeloid-derived sOPN also drives tissue remodeling by enhancing cell proliferation and metabolic activation. There are conflicting reports regarding sOPN’s impact on macrophage polarization. Some of them show an M1 polarization mediated by OPN in osteoarthritis [64], whereas other research studies describe how M2 polarization is promoted by OPN fragments. This process is the result of an enhanced MMP production that degrades OPN [65]. Another example of M2 polarization mediated by OPN is in lung adenocarcinoma via the upregulation of PD-L1 to enhance the metastatic process of the cancer [66].

3.1.6. Osteopontin as an Intracellular Molecule

Some cells like macrophages and dendritic cells also produce iOPN observed in the cytoplasm at resting conditions and relocating to the submembranous region where it activates its receptors after stimulation. During mitosis, iOPN is also detected diffusely in the nucleus [5].

3.1.7. Osteopontin in Non-Malignant Diseases

OPN is involved in wound healing resolution by increasing macrophage infiltration, transforming growth factor (TGF) β secretion, and fibroblast proliferation, and by modulating MMP expression [67]. In heart failure, myocardial infarction, and peripheral vascular disease, OPN possesses ambivalent functions. Its anti-inflammatory role limits cardiac fibrosis, vascular calcification, and cardiac tissue remodeling via modulation of collagen deposition and inducible nitric oxide synthase (iNOS) inhibition. On the contrary, the pro-inflammatory effects of OPN on the cardiovascular system include the enhancement of toxic nitric oxide metabolites, fibrosis, calcification, and tissue remodeling via chemoattraction of macrophages, endothelial cells, and smooth muscle cells [68]. In healthy lungs, OPN exhibits protective functions against injury by limiting potential toxicity from the neutrophil extracellular trap (NET) defense mechanism. In more detail, during NETosis, neutrophil DNA traps pathogens, thereby also releasing cytotoxic cationic histones. OPN with its anionic structure can bind these histones and limit their toxic effects [42].

3.2. Osteopontin Signaling in Inflammation

3.2.1. Inflammatory Signaling Pathways Initiated by Osteopontin

OPN is a key mediator of inflammatory responses mostly functioning as a chemoattractant for immune cells. The main role of OPN during inflammation is to trigger different leucocytes, induce cytokine secretion, and shape the immune response. At sites of injury, OPN can stimulate migration, accumulation, and persistence of macrophages and regulate their cytokine production by promoting Th1 cell-mediated immunity [69]. The interaction of OPN with α4 and α9 integrins and CD44 can promote either macrophage migration or apoptosis [70]. Wound healing studies have shown that OPN is highly expressed during the acute phase of inflammation and regulates tissue remodeling. Particularly in myocardial infarction, the IL-10-STAT3-galectin-3 axis is essential for OPN-producing reparative macrophage polarization that leads to tissue repair by enhancing fibrosis and clearance of apoptotic cells [71,72].
OPN participates in the pathogenesis of inflammatory and immune-mediated diseases and regulates the host response to infections [10]. For example, OPN promotes IL-12 secretion from dendritic cells (DCs) and their response to hepatitis B virus (HBV) infection, at the same time driving IFN-γ, IL-17A, and IL-10 secretion during Trypanosoma cruzi infection [31,73]. OPN also drives sepsis-induced acute lung injury by enhancing neutrophil infiltration via ERK and P38 MAPK activation [74]. Moreover, iOPN is implicated in the differentiation of long-lived natural killer (NK) cells with a memory-like phenotype following homeostatic expansion during host defense against pathogens or cancerous tumor formation. Its absence fails to maintain normal NK cellularity and its stimulation by IL-15 increases cell death [75]. However, OPN limits inflammation, tissue injury, and bacterial load during concurrent pneumococcal infection and allergic airway inflammation [76]. It can be upregulated together with leptin, and both of them may regulate apoptosis, adhesion, migration, and activation of eosinophils, in a PI3K and anti-α4 activation manner [77].
Both iOPN and sOPN control the relative abundance of myeloid and lymphoid cells. iOPN limits myeloid progenitor cell proliferation. E-cadherin induces iOPN which activates the Hippo pathway, thereby upregulating OPN mRNA in myeloid progenitors and eventually limiting their expansion. iOPN also exerts proapoptotic functions in the early stages of granulopoiesis. On the contrary, sOPN enhances lymphoid cell expansion through an anti-apoptotic mechanism [37].
OPN binds eosinophil-recruiting chemokines inhibiting their antibacterial activity against Streptococcus pneumoniae in parallel with sustained allergic inflammation during asthma [76].
OPN activity can also be annihilated by MMP cleavage in patients with autoimmune diseases, inhibiting OPN-driven cell adhesion and migration, and resulting in a less inflammatory phenotype [78].

3.2.2. Chronic Lung Diseases and Osteopontin

Several inflammatory diseases that are associated with airway remodeling depend on OPN signaling. During asthma, pulmonary fibrotic changes are mediated by interleukin-33 (IL-33)-enhanced amphiregulin production by memory T helper 2 cells. Amphiregulin along with epidermal growth factor receptor (EGFR)-signaling directly reprograms eosinophils to an inflammatory state and stimulates them to produce OPN, which acts as a pro-fibrotic mediator during the disease [79]. Arjomandi et al. also demonstrated that, in asthma, OPN is more fragmentated and can enhance the survival and the recruitment of alveolar macrophages [80]. In chronic obstructive pulmonary disease (COPD), OPN is upregulated and associated with exacerbations and may increase the vulnerability of the host defense to bacterial infections by interfering with the function of antimicrobial proteins [81,82,83]. Furthermore, OPN can be released by senescent pulmonary artery smooth muscle cells and contribute to the pathogenesis of pulmonary hypertension. OPN was recently reported to be positively correlated with plasma levels of IL-2 but not IL-4, which has anti-inflammatory effects in COPD and patients with both ischemic heart disease and increased pulmonary artery plasma pressure [84,85]. Although it is known that statins suppress OPN in cancer cells, their function in COPD was recently studied. Particularly, simvastatin was found to initiate downregulation of adenosine signaling which leads to direct inhibition of IL-13-activated STAT6 and finally repression of OPN expression, therefore preventing disease progression [86].
OPN has been also implicated in lung injury of different etiologies. Chronic exposure to carbon nanotubes (CNTs) that have been widely used in the last decades can cause serious injury in the lungs and lead to non-curable pulmonary fibrosis [87]. Lung interstitial fibrosis initiates with an acute inflammatory response, which is characterized by recruitment of inflammatory cells and secretion of pro-inflammatory and pro-fibrotic cytokines, chemokines, and growth factors, such as TNF-α, IL-1β, IL-6, MCP-1, and TGF-β1 [88]. After acute inflammatory and fibrotic responses, the pathologic effects transit to chronic inflammatory fibrosis evident by mild inflammation, increased deposition of ECM proteins, and formation of fibrotic foci [89]. The role of OPN was recently described in a mouse model of CNT exposure induced-fibrosis. It was reported that OPN is highly and persistently induced by CNTs in mouse lungs during both the acute and chronic phases of fibrosis and that OPN promotes lung fibrosis through the activation of TGF-β1 signaling and the promotion of myofibroblast differentiation [90]. TGF-β1 and OPN synergy have been also referred to as a possible target for inhibiting radiation-induced pulmonary fibrosis after lung cancer treatment [91]. Moreover, Fra-2 (a Fos member of the AP-1 transcription factor family) is linked to inflammation and fibrosis in the lungs. Particularly, in LPS-induced lung injury, Fra-2 recruits myeloid cells to the injured tissue in an OPN-depended manner, further exacerbating inflammation [92].
While there is still a need to better understand the pathogenesis of chronic lung scarring in idiopathic pulmonary fibrosis (IPF), studies have shown that OPN exerts severe pro-fibrotic effects [93]. White et al. described a panel of three proteins, namely, surfactant protein (SP)-D, matrix metalloproteinase (MMP)-7, and OPN, that can be used to distinguish IPF from other idiopathic interstitial lung diseases [94]. Additionally, a proteomic analysis of the BALF of patients with IPF has revealed strong expression of OPN along with upregulation of pro-fibrotic CCL24 [95].

3.3. Impact of Osteopontin on Cancer Development and Progression

OPN has been repeatedly implicated in the development and progression of several types of cancer. In most cancers, high expression levels of OPN or its isoforms are associated with poor prognosis (Table 2).

3.3.1. Impact of Osteopontin on Thoracic Cancers

Several lines of laboratory research, bioinformatics studies as well as meta-analyses have shown that OPN levels can also be considered as a biomarker for diagnosis or as a predicting tool for patient clinical outcome in several types of cancer, such as lung adenocarcinoma, esophageal carcinoma, prostate cancer, melanoma, and colorectal adenocarcinoma. Its expression levels are considered useful for predicting responses to anti-EGFR therapy in patients with triple-negative breast cancer and as a methylation-based biomarker for precise diagnosis and treatment for thyroid cancer [96,97,98,99,100,101,102].
Interestingly, OPN splice variants seem to differentially participate in cancer progression. For example, in thyroid cancer cells OPN-a overexpression stimulates higher matrix calcification and collagen synthesis when compared to overexpression of OPN-b or OPN-c [103]. In breast cancer, OPN-c is expressed in 75% of patients. At the same time, its expression is rising and is correlated with tumor stage in hepatocellular carcinoma (HCC) [104,105].

3.3.2. Osteopontin in Lung Cancer

As previously described, in most types of cancer, high levels of OPN are correlated with a more aggressive cancer phenotype and worse prognosis. OPN expression and activity in lung cancer is in accord with this phenotype (Figure 3). In this disease, OPN splice variants have been accused of differential role-playing. In NSCLC, OPN-a promotes a more aggressive phenotype due to the transcription of exon 4 in contrast to OPN-c, where the transcription of exon 4 is absent and is also demonstrated to promote the malignant phenotype of lung tumor cells by enhancing cell-adherent properties to bone tissues [106,107]. OPN-b induces lung cancer cell proliferation, whereas OPN-c enhances their invasive behavior [108]. Investigators have also focused on studying OPN gene single nucleotide polymorphisms (SNPs). These are characterized for their implementation in several types of cancer, but SNPs located in the promoter region of OPN have received investigators’ main focus. While rs11730582 is the most studied SNP of OPN with the −443 CC genotype generally associated with higher expression of OPN and increased cancer risk, this SNP tends to have a better prognosis in NSCLC. On the other hand, the SNP rs17524488 (−156G/G) is associated with higher levels of OPN expression and is correlated to a more advanced stage disease [109,110,111].
In patients with NSCLC, Sun et al. have reported that the combination of OPN and CD44v6 elevated expression of is a valuable independent predictor of tumor recurrence and survival in NSCLC patients [112]. For the same cancer, another study also demonstrated that VEGF and OPN are both overexpressed and positively correlated with tumor progression [12]. In particular, OPN stimulates autocrine and paracrine mechanisms which lead to VEGF accumulation, subsequently promoting tumorigenesis and metastasis [113]. During carcinogenesis, OPN upregulates PD-L1 and M2-polarized tumor-associated macrophages and facilitates tumor immune escape, further enhancing pro-tumor activity [66]. It promotes cancer cell drug resistance and invasion in patients with advanced NSCLC and facilitates the development of small-cell lung cancer (SCLC) cells by inhibiting apoptosis and autophagy [114,115]. Recent studies suggest that fibrocytes that are differentiated from bone marrow-derived CD14+ monocytes have features of both macrophages and fibroblasts and may promote cancer progression. They enhance the cancer-stem-cell-like properties of lung cancer cells through the secretion of OPN and its function in the activation of the PIK3K/AKT pathway [116].
Interestingly, lung cancers presenting genomic alterations leading to KRAS activation are characterized by resistance to radiation and poor prognosis. OPN and EGFR act in a synergistic way to initiate stem-like properties of tumor subpopulations whose characteristics are mitosis-like condensed chromatin (MLCC) and high expression of CD133 [117]. In another study, where the transmembrane 4 L6 family member 4 (TM4SF4) known to promote cancer was studied, authors demonstrated that OPN induced by upregulated β-catenin via TM4SF4-driven phosphorylation of glycogen synthase kinase 3b (GSK3β) activated the JAK2/STAT3 or FAK/STAT3 pathway in human lung cancer cells, which also upregulates OPN expression in an autocrine manner [118]. In SCLC patients, OPN has been reported to be one of the most upregulated genes and its inhibition can prevent epithelial–mesenchymal transition (EMT) formation, rendering it a prognostic tool for disease progression [117]. Finally, in patients with lung adenocarcinoma, OPN was identified as a biomarker, and high expression of the protein has been associated with poor patient survival and identified as a predicting tool for disease stage prognosis [96,119].

3.3.3. Malignant Pleural Effusion Promotion by Osteopontin

Malignant pleural effusion (MPE) is a complication of advanced lung, breast, pleural, and other cancers. In most studies performed in the past, enzyme-linked immunosorbent assay (ELISA) and correlations with progression-free survival or overall survival have been used to define OPN expression levels in patients with MPE, emphasizing OPN’s critical role in prognosis [120,121]. Nevertheless, Hsu et al. showed very recently that although pleural fluid OPN along with VEGF and uPA concentrations are elevated in MPE, they are not satisfactory predictors of pleurodesis outcome or patient survival [122]. Moreover, during tumor formation and the interaction of tumor cells with inflammatory responses, mast cells (like several other inflammatory cells) are reported to be recruited in lung adenocarcinomas and interestingly in MPE formation [123]. During these processes, tumor-derived OPN has been reported to promote the pleural fluid formation and pleural tumor progression by inducing vascular hyperpermeability and was recently further described to promote the activation and degranulation of mast cells in the pleural space [123,124]. MPE is a condition that also accompanies malignant pleural mesothelioma (MPM) progression mainly in asbestos-exposed industry workers. Elevated OPN expression levels have been reported during disease progression which is inhibited by zoledronic acid as a single agent in the treatment of MPM [125,126,127,128]. OPN has also been reported to be a target for pemetrexed-based chemotherapy in MPM cells, and its inhibition is accompanied by a reduction in the phosphorylation of AKT [129].

3.3.4. Osteopontin as a Pro-Metastatic Molecule

The mechanisms underlying the aggressive and invasive phenotype of tumor cells associated with OPN expression have been extensively investigated. Although OPN gene silencing has been reported in a study by Sun et al. [130] to contribute to lung tumor cell metastasis by stimulating cell invasion and not cellular migration and proliferation, Polat et al. demonstrated in another study that lung cancer cell proliferation and migration were significantly reduced when OPN was silenced [131]. Moreover, OPN overexpression is shown to activate the PI3K-AKT-Twist pathway, thus promoting EMT and resulting in colorectal cancer metastases [132]. OPN exerts its function by inducing changes in EMT and promoting the initiation of metastases in different types of cancer. OPN can cause an increase in EMT-related transcription factors including Twist, Snail, and Slug [133]. Its overexpression results in serine phosphorylation of Twist, which then binds to the Bmi-1 promoter, subsequently activating EMT in breast cancer cells [134]. Furthermore, OPN can promote EMT-mediated metastases by inducing hypoxia-inducible factor-1 alpha (HIF-1α). Intra-tumor hypoxia stabilizes HIF-1α, which regulates the expression of Twist by binding to the Twist promoter, thus inducing EMT. OPN was shown to increase HIF-1α via the PI3k/AKT pathway in a breast cancer model and induce EMT of HCC cells via increasing vimentin stability, leading to the promotion of HCC metastasis [135,136]. Moreover, a recent study including experiments on liver and lung cancer cells revealed that iOPN translocates to the nucleus and induces mesenchymal-to-epithelial transition (MET) to facilitate the formation of metastases. In particular, nuclear OPN was found to interact with HIF-2α and impact the AKT1/miR-429/ZEB cascade, subsequently enhancing the progression of metastatic colonization. The translocation of OPN into the nucleus is dependent on micro-environmental VEGF activity through a KDR/PLCγ/PKC-dependent pathway [137]. In NSCLC metastasis, OPN plays a key role by inducing cancer-associated fibroblasts (CAFs) [133] and can induce tumor cell migration by interacting with integrins and CD44 [138].
OPN isoforms can differentially facilitate lung metastasis. Tumor-originated iOPN may promote tumor cell survival by preventing tumor-related protein 53 (TP53)-mediated apoptosis, whereas tumor-derived sOPN may function in a paracrine mode to accelerate lung metastasis by enhancing tumor-derived CCL2 chemokine signaling to cognate host receptors [139]. Additionally, MMP-9 activity can lead to an OPN-32 kDa cleaved isoform that induces the production of myeloid-derived suppressor cells, thus mediating tumor immune escape [140]. OPN may also activate ROCK signaling via the FAK/PI3K/AKT pathway, thereby facilitating the invasion of lung cancer cells through lamellipodia formation and the inactivation of cofilin [141].
Finally, lysosomal-associated membrane protein 3 (LAMP3) is a tumor-specific protein induced by hypoxia, which stimulates invasion and metastasis of various cancer cells and has been described to possibly inhibit OPN function by regulating downstream signaling of OPN in osteosarcoma metastasis in the lungs [142]. In another metastatic tumor model, where 4T1 mammary tumor cells are injected into mice, OPN was demonstrated to be required for activating ERK in polymorphonuclear cells in the tumor emboli site and to facilitate tumor colonization through NETosis [143].

3.4. Potential Clinical Implications for Osteopontin in Cancer

3.4.1. Osteopontin as a Biomarker

Circulating OPN levels are elevated in several cancers, including mesothelioma [144], colon [145], lung [146], and breast cancer [147]. In malignant mesothelioma (MM), OPN is considered as a potential biomarker and has been quantified in the serum to detect early stages of the disease [148]. More specifically, Pass et al. determined the correlation between MM and increased serum OPN concentrations. They quantified OPN levels in the serum and the tumor tissues in 49 control patients (without any documented exposure to asbestos), 69 asbestos-exposed subjects, and 76 patients with MM [148]. That study showed that patients with or without asbestos exposure had similar OPN levels in the serum. However, in MM, serum OPN concentrations were significantly elevated compared to controls and asbestos-exposed subjects. The serum OPN assay performed by Pass et al. presented a sensitivity of 77.6% and a specificity of 85.5%, denoting OPN as a promising biomarker of MM identification in clinical routine [148]. Other studies revealed conflicting results in the diagnostic efficiency of MM via circulating OPN levels [149,150,151,152,153]. Some of the discrepancies can be interpreted by the origin of the samples used for the OPN assay. The overall diagnostic accuracy of serum OPN to diagnose MM was assessed in a recent systematic review with meta-analysis [150]. That study reported that pooled sensitivity was 57% (95% CI 52–61%), and specificity was 81% (95% CI 79–84%). Moreover, the area under the curve (AUC) of the receiver operating characteristic (ROC) curve was equal to 0.80. Altogether, these results suggest that OPN is a helpful biomarker for MM diagnosis [150].
Contradictory studies exist regarding lung cancer stage and OPN levels. Some studies describe that advanced stages of NSCLC present higher levels of serum OPN compared to the early stages [111]. However, other studies did not detect a significant difference in OPN concentrations according to TNM classification [154,155]. In those studies, the limited number of patients with early-stage disease may be the cause of a lack of statistical significance, which is more compatible with what is observed in daily clinical practice. Other differences could be explained by the tests used for quantification, as well as by thrombin cleavage of OPN during clotting with the consequence of a lower quantifiable OPN amount in serum compared to plasma [154]. Currently, OPN is not an established prognostic or diagnostic biomarker for cancer, and further studies to assess the value of OPN as a biomarker are needed.

3.4.2. Osteopontin as a Therapeutic Target

There are currently no clinical trials targeting OPN as a treatment for lung cancer or mesothelioma (latest review of https://clinicaltrials.gov/ on 1 May 2019). Currently, two studies are recruiting and aim to assess the role of OPN as a biomarker of arteriovenous fistula (NCT03270358) without stenosis and as a risk factor for cerebrovascular stroke (NCT03561285). The only relevant trials are cohort/database studies focused on mesothelioma and aimed at exploring the utility of OPN as a biomarker for mesothelioma.

3.4.3. Tools for Future Research on Osteopontin

In order to study and target OPN, different tools are already available (Table 3). On the one hand, there are tools to produce and functionally analyze OPN truncations. The expression vectors are useful for the overexpression of the protein or the introduction of mutations, while knockout mice models such as the B6.129S6(Cg)-Spp1tm1Blh/J strain [156] are animals without functional OPN. All these tools are allowing the study of OPN influence over organisms or diseases like lung cancer or mesothelioma.
On the other hand, siRNA strategies have also been developed to inhibit OPN. For instance, Zhao et al. showed that a lentivirus-mediated RNAi can reduce the proliferation of the human lung cancer cell line A549 [108]. Blocking antibodies and small molecule inhibitors are also available to neutralize OPN activity. As an example, brefelamide suppresses the in vitro invasion capability of A549 lung cancer cells by inhibiting OPN [14]. The human antibody AOM1 is recognizing the epitope SVVYGLRSKS in human and murine OPN, inhibiting its capability of binding integrins, thereby decreasing the growth of metastatic tumors in a model of non-small lung cancer [13].

4. Conclusions and Outlook

Taken together, OPN and its variants are highly involved in the regulation of tumor-associated inflammation as well as the aggressiveness of cancer cells and tumor growth enhancement. Although there are still unknown mechanisms of action of each OPN isoform, it is obvious that OPN variants may be interesting therapeutic targets for chronic inflammatory diseases and cancer. OPN-based clinical tests could be developed to evaluate disease prognosis and response to therapy. Indeed, recent breakthroughs in the targeting of OPN and in the development of tools to analyze the protein are exciting. It is anticipated that soon, new strategies will be developed and significant advancements will be made along these fronts.

Author Contributions

Conceptualization, G.T.S. and A.-S.L; writing—original draft preparation, A.-S.L., I.G., I.P. and G.T.S.; writing—review and editing, G.T.S. and A.-S.L.; visualization, A.-S.L.; supervision, G.T.S.

Funding

This work was supported by institutional funds. The study sponsors had no role in study design, data collection, analysis, or interpretation, in writing, or in submitting the paper for publication.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Young, M.F.; Kerr, J.M.; Termine, J.D.; Wewer, U.M.; Wang, M.G.; McBride, O.W.; Fisher, L.W. cDNA cloning, mRNA distribution and heterogeneity, chromosomal location, and RFLP analysis of human osteopontin (OPN). Genomics 1990, 7, 491–502. [Google Scholar] [CrossRef] [Green Version]
  2. Fet, V.; Dickinson, M.E.; Hogan, B.L.M. Localization of the mouse gene for secreted phosphoprotein 1 (Spp-1) (2ar, osteopontin, bone sialoprotein 1, 44-kDa bone phosphoprotein, tumor-secreted phosphoprotein) to chromosome 5, closely linked to Ric (Rickettsia resistance). Genomics 1989, 5, 375–377. [Google Scholar] [CrossRef]
  3. Chae, S.; Jun, H.-O.; Lee, E.G.; Yang, S.-J.; Lee, D.C.; Jung, J.K.; Park, K.C.; Yeom, Y.I.; Kim, K.-W. Osteopontin splice variants differentially modulate the migratory activity of hepatocellular carcinoma cell lines. Int. J. Oncol. 2009, 35, 1409–1416. [Google Scholar] [Green Version]
  4. Standal, T.; Borset, M.; Sundan, A. Role of osteopontin in adhesion, migration, cell survival and bone remodeling. Exp. Oncol. 2004, 26, 179–184. [Google Scholar]
  5. Inoue, M.; Shinohara, M.L. Intracellular osteopontin (iOPN) and immunity. Immunol. Res. 2011, 49, 160–172. [Google Scholar] [CrossRef]
  6. Christensen, B.; Nielsen, M.S.; Haselmann, K.F.; Petersen, T.E.; Sørensen, E.S. Post-translationally modified residues of native human osteopontin are located in clusters: Identification of 36 phosphorylation and five O-glycosylation sites and their biological implications. Biochem. J. 2005, 390, 285–292. [Google Scholar] [CrossRef]
  7. Li, H.; Shen, H.; Yan, G.; Zhang, Y.; Liu, M.; Fang, P.; Yu, H.; Yang, P. Site-specific structural characterization of O-glycosylation and identification of phosphorylation sites of recombinant osteopontin. Biochimica Biophysica Acta (BBA)—Proteins Proteom. 2015, 1854, 581–591. [Google Scholar] [CrossRef]
  8. Maeda, N.; Maenaka, K. The Roles of Matricellular Proteins in Oncogenic Virus-Induced Cancers and Their Potential Utilities as Therapeutic Targets. Int. J. Mol. Sci. 2017, 18, 2198. [Google Scholar] [CrossRef]
  9. McKee, M.D.; Pedraza, C.E.; Kaartinen, M.T. Osteopontin and wound healing in bone. Cells Tissues Organs (Print) 2011, 194, 313–319. [Google Scholar] [CrossRef]
  10. Rittling, S.R.; Singh, R. Osteopontin in Immune-mediated Diseases. J. Dent. Res. 2015, 94, 1638–1645. [Google Scholar] [CrossRef] [Green Version]
  11. Klement, J.D.; Paschall, A.V.; Redd, P.S.; Ibrahim, M.L.; Lu, C.; Yang, D.; Celis, E.; Abrams, S.I.; Ozato, K.; Liu, K. An osteopontin/CD44 immune checkpoint controls CD8+ T cell activation and tumor immune evasion. J. Clin. Investig. 2018, 128, 5549–5560. [Google Scholar] [CrossRef]
  12. Lin, Q.; Guo, L.; Lin, G.; Chen, Z.; Chen, T.; Lin, J.; Zhang, B.; Gu, X. Clinical and prognostic significance of OPN and VEGF expression in patients with non-small-cell lung cancer. Cancer Epidemiol. 2015, 39, 539–544. [Google Scholar] [CrossRef]
  13. Shojaei, F.; Scott, N.; Kang, X.; Lappin, P.B.; Fitzgerald, A.A.; Karlicek, S.; Simmons, B.H.; Wu, A.; Lee, J.H.; Bergqvist, S.; et al. Osteopontin induces growth of metastatic tumors in a preclinical model of non-small lung cancer. J. Exp. Clin. Cancer Res. 2012, 31, 26. [Google Scholar] [CrossRef]
  14. Zhang, J.; Yamada, O.; Kida, S.; Matsushita, Y.; Murase, S.; Hattori, T.; Kubohara, Y.; Kikuchi, H.; Oshima, Y. Identification of brefelamide as a novel inhibitor of osteopontin that suppresses invasion of A549 lung cancer cells. Oncol. Rep. 2016, 36, 2357–2364. [Google Scholar] [CrossRef]
  15. Senger, D.R.; Wirth, D.F.; Hynes, R.O. Transformed mammalian cells secrete specific proteins and phosphoproteins. Cell 1979, 16, 885–893. [Google Scholar] [CrossRef]
  16. Franzén, A.; Heinegård, D. Isolation and characterization of two sialoproteins present only in bone calcified matrix. Biochem. J. 1985, 232, 715–724. [Google Scholar] [CrossRef] [Green Version]
  17. Oldberg, A.; Franzén, A.; Heinegård, D. Cloning and sequence analysis of rat bone sialoprotein (osteopontin) cDNA reveals an Arg-Gly-Asp cell-binding sequence. Proc. Natl. Acad. Sci. USA 1986, 83, 8819–8823. [Google Scholar] [CrossRef]
  18. Fisher, L.W.; Torchia, D.A.; Fohr, B.; Young, M.F.; Fedarko, N.S. Flexible Structures of SIBLING Proteins, Bone Sialoprotein, and Osteopontin. Biochem. Biophys. Res. Commun. 2001, 280, 460–465. [Google Scholar] [CrossRef] [Green Version]
  19. Briones-Orta, M.A.; Avendaño-Vázquez, S.E.; Aparicio-Bautista, D.I.; Coombes, J.D.; Weber, G.F.; Syn, W.-K. Osteopontin splice variants and polymorphisms in cancer progression and prognosis. Biochim. Biophys. Acta Rev. Cancer 2017, 1868, 93–108. [Google Scholar] [CrossRef]
  20. Hijiya, N.; Setoguchi, M.; Matsuura, K.; Higuchi, Y.; Akizuki, S.; Yamamoto, S. Cloning and characterization of the human osteopontin gene and its promoter. Biochem. J. 1994, 303, 255–262. [Google Scholar] [CrossRef] [Green Version]
  21. He, B.; Mirza, M.; Weber, G.F. An osteopontin splice variant induces anchorage independence in human breast cancer cells. Oncogene 2006, 25, 2192–2202. [Google Scholar] [CrossRef]
  22. Carvalho, M.S.; Cabral, J.M.; da Silva, C.L.; Vashishth, D. Synergistic effect of extracellularly supplemented osteopontin and osteocalcin on stem cell proliferation, osteogenic differentiation, and angiogenic properties. J. Cell. Biochem. 2018, 120, 6555–6569. [Google Scholar] [CrossRef]
  23. SPP1 (Secreted Phosphoprotein 1). Available online: http://atlasgeneticsoncology.org/Genes/SPP1ID42379ch4q22 (accessed on 5 December 2018).
  24. Sodek, J.; Chen, J.; Nagata, T.; Kasugai, S.; Todescan, R.; Li, I.W.; Kim, R.H. Regulation of osteopontin expression in osteoblasts. Ann. N. Y. Acad. Sci. 1995, 760, 223–241. [Google Scholar] [CrossRef]
  25. Zohar, R.; Lee, W.; Arora, P.; Cheifetz, S.; McCulloch, C.; Sodek, J. Single cell analysis of intracellular osteopontin in osteogenic cultures of fetal rat calvarial cells. J. Cell. Physiol. 1997, 170, 88–100. [Google Scholar] [CrossRef]
  26. Ikegame, M.; Ejiri, S.; Okamura, H. Expression of Noncollagenous Bone Matrix Proteins in Osteoblasts Stimulated by Mechanical Stretching in the Cranial Suture of Neonatal Mice—PubMed-NCBI. Available online: https://www.ncbi.nlm.nih.gov/pubmed/30113872 (accessed on 2 January 2019).
  27. Teixeira, L.N.; de Castro Raucci, L.M.S.; Alonso, G.C.; Coletta, R.D.; Rosa, A.L.; de Oliveira, P.T. Osteopontin expression in co-cultures of human squamous cell carcinoma-derived cells and osteoblastic cells and its effects on the neoplastic cell phenotype and osteoclastic activation. Tumour Biol. 2016, 37, 12371–12385. [Google Scholar] [CrossRef]
  28. Qian, J.; Xu, L.; Sun, X.; Wang, Y.; Xuan, W.; Zhang, Q.; Zhao, P.; Wu, Q.; Liu, R.; Che, N.; et al. Adiponectin aggravates bone erosion by promoting osteopontin production in synovial tissue of rheumatoid arthritis. Arthritis Res. Ther. 2018, 20, 26. [Google Scholar] [CrossRef] [Green Version]
  29. Pestell, T.G.; Jiao, X.; Kumar, M.; Peck, A.R.; Prisco, M.; Deng, S.; Li, Z.; Ertel, A.; Casimiro, M.C.; Ju, X.; et al. Stromal cyclin D1 promotes heterotypic immune signaling and breast cancer growth. Oncotarget 2017, 8, 81754–81775. [Google Scholar] [CrossRef]
  30. Kaleta, B.; Boguska, A. Sildenafil, a Phosphodiesterase Type 5 Inhibitor, Downregulates Osteopontin in Human Peripheral Blood Mononuclear Cells. Arch. Immunol. Ther. Exp. (Warsz.) 2017, 65, 347–353. [Google Scholar] [CrossRef] [Green Version]
  31. Cui, G.; Chen, J.; He, J.; Lu, C.; Wei, Y.; Wang, L.; Xu, X.; Li, L.; Uede, T.; Diao, H. Osteopontin promotes dendritic cell maturation and function in response to HBV antigens. Drug Des. Dev. Ther. 2015, 9, 3003–3016. [Google Scholar]
  32. Ahmad, R.; Al-Mass, A.; Al-Ghawas, D.; Shareif, N.; Zghoul, N.; Melhem, M.; Hasan, A.; Al-Ghimlas, F.; Dermime, S.; Behbehani, K. Interaction of osteopontin with IL-18 in obese individuals: Implications for insulin resistance. PLoS ONE 2013, 8, e63944. [Google Scholar] [CrossRef]
  33. Shinohara, M.L.; Jansson, M.; Hwang, E.S.; Werneck, M.B.F.; Glimcher, L.H.; Cantor, H. T-bet-dependent expression of osteopontin contributes to T cell polarization. Proc. Natl. Acad. Sci. USA 2005, 102, 17101–17106. [Google Scholar] [CrossRef] [Green Version]
  34. Vaschetto, R.; Nicola, S.; Olivieri, C.; Boggio, E.; Piccolella, F.; Mesturini, R.; Damnotti, F.; Colombo, D.; Navalesi, P.; Della Corte, F.; et al. Serum levels of osteopontin are increased in SIRS and sepsis. Intensive Care Med. 2008, 34, 2176–2184. [Google Scholar] [CrossRef]
  35. Schack, L.; Lange, A.; Kelsen, J.; Agnholt, J.; Christensen, B.; Petersen, T.E.; Sørensen, E.S. Considerable variation in the concentration of osteopontin in human milk, bovine milk, and infant formulas. J. Dairy Sci. 2009, 92, 5378–5385. [Google Scholar] [CrossRef]
  36. Kolbach, A.M.; Afzal, O.; Halligan, B.; Sorokina, E.; Kleinman, J.G.; Wesson, J.A. Relative deficiency of acidic isoforms of osteopontin from stone former urine. Urol. Res. 2012, 40, 447–454. [Google Scholar] [CrossRef]
  37. Kanayama, M.; Xu, S.; Danzaki, K.; Gibson, J.R.; Inoue, M.; Gregory, S.G.; Shinohara, M.L. Skewing of the population balance of lymphoid and myeloid cells by secreted and intracellular osteopontin. Nat. Immunol. 2017, 18, 973–984. [Google Scholar] [CrossRef] [Green Version]
  38. Inoue, M.; Arikawa, T.; Chen, Y.-H.; Moriwaki, Y.; Price, M.; Brown, M.; Perfect, J.R.; Shinohara, M.L. T cells down-regulate macrophage TNF production by IRAK1-mediated IL-10 expression and control innate hyperinflammation. Proc. Natl. Acad. Sci. USA 2014, 111, 5295–5300. [Google Scholar] [CrossRef] [Green Version]
  39. Shinohara, M.L.; Kim, H.-J.; Kim, J.-H.; Garcia, V.A.; Cantor, H. Alternative translation of osteopontin generates intracellular and secreted isoforms that mediate distinct biological activities in dendritic cells. Proc. Natl. Acad. Sci. USA 2008, 105, 7235–7239. [Google Scholar] [CrossRef] [Green Version]
  40. Jovic, S.; Shikhagaie, M.; Mörgelin, M.; Erjefält, J.S.; Kjellström, S.; Egesten, A. Osteopontin is increased in cystic fibrosis and can skew the functional balance between ELR-positive and ELR-negative CXC-chemokines. J. Cyst. Fibros. 2015, 14, 453–463. [Google Scholar] [CrossRef] [Green Version]
  41. Samitas, K.; Zervas, E.; Vittorakis, S.; Semitekolou, M.; Alissafi, T.; Bossios, A.; Gogos, H.; Economidou, E.; Lötvall, J.; Xanthou, G.; et al. Osteopontin expression and relation to disease severity in human asthma. Eur. Respir. J. 2011, 37, 331–341. [Google Scholar] [CrossRef]
  42. Kasetty, G.; Papareddy, P.; Bhongir, R.K.V.; Ali, M.N.; Mori, M.; Wygrecka, M.; Erjefält, J.S.; Hultgårdh-Nilsson, A.; Palmberg, L.; Herwald, H.; et al. Osteopontin protects against lung injury caused by extracellular histones. Mucosal Immunol. 2019, 12, 39. [Google Scholar] [CrossRef]
  43. Minai-Tehrani, A.; Chang, S.-H.; Park, S.B.; Cho, M.-H. The O-glycosylation mutant osteopontin alters lung cancer cell growth and migration in vitro and in vivo. Int. J. Mol. Med. 2013, 32, 1137–1149. [Google Scholar] [CrossRef] [Green Version]
  44. Liu, Y.-N.; Kang, B.-B.; Chen, J.H. Transcriptional regulation of human osteopontin promoter by C/EBPalpha and AML-1 in metastatic cancer cells. Oncogene 2004, 23, 278–288. [Google Scholar] [CrossRef]
  45. Takami, Y.; Russell, M.B.; Gao, C.; Mi, Z.; Guo, H.; Mantyh, C.R.; Kuo, P.C. Sp1 regulates osteopontin expression in SW480 human colon adenocarcinoma cells. Surgery 2007, 142, 163–169. [Google Scholar] [CrossRef] [Green Version]
  46. Zhang, W.; Li, Q.; Li, D.; Li, J.; Aki, D.; Liu, Y.-C. The E3 ligase VHL controls alveolar macrophage function via metabolic-epigenetic regulation. J. Exp. Med. 2018, 215, 3180–3193. [Google Scholar] [CrossRef]
  47. Zhang, J.; Yamada, O.; Kida, S.; Matsushita, Y.; Hattori, T. Down-regulation of osteopontin mediates a novel mechanism underlying the cytostatic activity of TGF-β. Cell. Oncol. 2016, 39, 119–128. [Google Scholar] [CrossRef]
  48. Schultz, J.; Lorenz, P.; Ibrahim, S.M.; Kundt, G.; Gross, G.; Kunz, M. The functional-443T/C osteopontin promoter polymorphism influences osteopontin gene expression in melanoma cells via binding of c-Myb transcription factor. Mol. Carcinog. 2009, 48, 14–23. [Google Scholar] [CrossRef]
  49. Flajollet, S.; Tian, T.V.; Flourens, A.; Tomavo, N.; Villers, A.; Bonnelye, E.; Aubert, S.; Leroy, X.; Duterque-Coquillaud, M. Abnormal expression of the ERG transcription factor in prostate cancer cells activates osteopontin. Mol. Cancer Res. 2011, 9, 914–924. [Google Scholar] [CrossRef]
  50. Yang, Y.; Yang, Y.; Li, X.; Cui, L.; Fu, M.; Rabie, A.B.; Zhang, D. Functional analysis of core binding factor a1 and its relationship with related genes expressed by human periodontal ligament cells exposed to mechanical stress. Eur. J. Orthod. 2010, 32, 698–705. [Google Scholar] [CrossRef] [Green Version]
  51. Guarneri, C.; Bevelacqua, V.; Polesel, J.; Falzone, L.; Cannavò, P.S.; Spandidos, D.A.; Malaponte, G.; Libra, M. NF-κB inhibition is associated with OPN/MMP-9 downregulation in cutaneous melanoma. Oncol. Rep. 2017, 37, 737–746. [Google Scholar] [CrossRef]
  52. Hu, B.; Zhou, H.; Gao, H.; Liu, Y.; Yan, T.; Zou, L.; Chen, L. IFN-γ inhibits osteopontin expression in human decidual stromal cells and can be attenuated by 1α,25-dihydroxyvitamin D3. Am. J. Reprod. Immunol. 2012, 68, 353–361. [Google Scholar] [CrossRef]
  53. Cai, M.; Bompada, P.; Salehi, A.; Acosta, J.R.; Prasad, R.B.; Atac, D.; Laakso, M.; Groop, L.; De Marinis, Y. Role of osteopontin and its regulation in pancreatic islet. Biochem. Biophys. Res. Commun. 2018, 495, 1426–1431. [Google Scholar] [CrossRef]
  54. Liu, W.-L.; Zhang, H.; Zheng, Y.; Wang, H.-T.; Chen, F.-H.; Xu, L.; Wei, Y.; Sun, Y.-Q.; Shi, J.-B.; Li, H.-B. Expression and regulation of osteopontin in chronic rhinosinusitis with nasal polyps. Clin. Exp. Allergy 2015, 45, 414–422. [Google Scholar] [CrossRef]
  55. Chen, M.; Chen, G.; Nie, H.; Zhang, X.; Niu, X.; Zang, Y.C.Q.; Skinner, S.M.; Zhang, J.Z.; Killian, J.M.; Hong, J. Regulatory effects of IFN-beta on production of osteopontin and IL-17 by CD4+ T Cells in MS. Eur. J. Immunol. 2009, 39, 2525–2536. [Google Scholar] [CrossRef]
  56. Scutera, S.; Salvi, V.; Lorenzi, L.; Piersigilli, G.; Lonardi, S.; Alotto, D.; Casarin, S.; Castagnoli, C.; Dander, E.; D’Amico, G.; et al. Adaptive Regulation of Osteopontin Production by Dendritic Cells Through the Bidirectional Interaction with Mesenchymal Stromal Cells. Front. Immunol. 2018, 9, 1207. [Google Scholar] [CrossRef]
  57. Lin, Y.; McKinnon, K.E.; Ha, S.W.; Beck, G.R. Inorganic phosphate induces cancer cell mediated angiogenesis dependent on forkhead box protein C2 (FOXC2) regulated osteopontin expression. Mol. Carcinog. 2015, 54, 926–934. [Google Scholar] [CrossRef]
  58. Labudzynskyi, D.; Shymanskyy, I.; Veliky, M. Role of vitamin D3 in regulation of interleukin-6 and osteopontin expression in liver of diabetic mice. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 2916–2919. [Google Scholar]
  59. Zhuang, S.; Hua, X.; He, K.; Zhou, T.; Zhang, J.; Wu, H.; Ma, X.; Xia, Q.; Zhang, J. Inhibition of GSK-3β induces AP-1-mediated osteopontin expression to promote cholestatic liver fibrosis. FASEB J. 2018, 32, 4494–4503. [Google Scholar] [CrossRef]
  60. Giachelli, C.M.; Steitz, S. Osteopontin: A versatile regulator of inflammation and biomineralization. Matrix Biol. 2000, 19, 615–622. [Google Scholar] [CrossRef]
  61. Agnihotri, R.; Crawford, H.C.; Haro, H.; Matrisian, L.M.; Havrda, M.C.; Liaw, L. Osteopontin, a Novel Substrate for Matrix Metalloproteinase-3 (Stromelysin-1) and Matrix Metalloproteinase-7 (Matrilysin). J. Biol. Chem. 2001, 276, 28261–28267. [Google Scholar] [CrossRef] [Green Version]
  62. Sun, S.-J.; Wu, C.-C.; Sheu, G.-T.; Chang, H.-Y.; Chen, M.-Y.; Lin, Y.-Y.; Chuang, C.-Y.; Hsu, S.-L.; Chang, J.T. Integrin β3 and CD44 levels determine the effects of the OPN-a splicing variant on lung cancer cell growth. Oncotarget 2016, 7, 55572–55584. [Google Scholar] [CrossRef]
  63. Dai, J.; Peng, L.; Fan, K.; Wang, H.; Wei, R.; Ji, G.; Cai, J.; Lu, B.; Li, B.; Zhang, D.; et al. Osteopontin induces angiogenesis through activation of PI3K/AKT and ERK1/2 in endothelial cells. Oncogene 2009, 28, 3412–3422. [Google Scholar] [CrossRef] [Green Version]
  64. Li, L.; Lv, G.; Wang, B.; Kuang, L. XIST/miR-376c-5p/OPN axis modulates the influence of proinflammatory M1 macrophages on osteoarthritis chondrocyte apoptosis. J. Cell. Physiol. 2019. [Google Scholar] [CrossRef]
  65. Ge, Q.; Ruan, C.-C.; Ma, Y.; Tang, X.-F.; Wu, Q.-H.; Wang, J.-G.; Zhu, D.-L.; Gao, P.-J. Osteopontin regulates macrophage activation and osteoclast formation in hypertensive patients with vascular calcification. Sci. Rep. 2017, 7, 40253. [Google Scholar] [CrossRef] [Green Version]
  66. Zhang, Y.; Du, W.; Chen, Z.; Xiang, C. Upregulation of PD-L1 by SPP1 mediates macrophage polarization and facilitates immune escape in lung adenocarcinoma. Exp. Cell Res. 2017, 359, 449–457. [Google Scholar] [CrossRef]
  67. Icer, M.A.; Gezmen-Karadag, M. The multiple functions and mechanisms of osteopontin. Clin. Biochem. 2018, 59, 17–24. [Google Scholar] [CrossRef]
  68. Waller, A.H.; Sanchez-Ross, M.; Kaluski, E.; Klapholz, M. Osteopontin in cardiovascular disease: A potential therapeutic target. Cardiol. Rev. 2010, 18, 125–131. [Google Scholar] [CrossRef]
  69. Kahles, F.; Findeisen, H.M.; Bruemmer, D. Osteopontin: A novel regulator at the cross roads of inflammation, obesity and diabetes. Mol. Metab. 2014, 3, 384–393. [Google Scholar] [CrossRef]
  70. Lund, S.A.; Wilson, C.L.; Raines, E.W.; Tang, J.; Giachelli, C.M.; Scatena, M. Osteopontin mediates macrophage chemotaxis via α4 and α9 integrins and survival via the α4 integrin. J. Cell. Biochem. 2013, 114, 1194–1202. [Google Scholar] [CrossRef]
  71. Shirakawa, K.; Endo, J.; Kataoka, M.; Katsumata, Y.; Yoshida, N.; Yamamoto, T.; Isobe, S.; Moriyama, H.; Goto, S.; Kitakata, H.; et al. IL (Interleukin)-10-STAT3-Galectin-3 Axis Is Essential for Osteopontin-Producing Reparative Macrophage Polarization After Myocardial Infarction. Circulation 2018, 138, 2021–2035. [Google Scholar] [CrossRef]
  72. Chen, J.; Yang, L.; Chen, Y.; Zhang, G.; Fan, Z. Controlled release of osteopontin and interleukin-10 from modified endovascular coil promote cerebral aneurysm healing. J. Neurol. Sci. 2016, 360, 13–17. [Google Scholar] [CrossRef]
  73. Santamaría, M.H.; Corral, R.S. Osteopontin-dependent regulation of Th1 and Th17 cytokine responses in Trypanosoma cruzi-infected C57BL/6 mice. Cytokine 2013, 61, 491–498. [Google Scholar] [CrossRef]
  74. Hirano, Y.; Aziz, M.; Yang, W.-L.; Wang, Z.; Zhou, M.; Ochani, M.; Khader, A.; Wang, P. Neutralization of osteopontin attenuates neutrophil migration in sepsis-induced acute lung injury. Crit. Care 2015, 19, 53. [Google Scholar] [CrossRef]
  75. Leavenworth, J.W.; Verbinnen, B.; Wang, Q.; Shen, E.; Cantor, H. Intracellular osteopontin regulates homeostasis and function of natural killer cells. Proc. Natl. Acad. Sci. USA 2015, 112, 494–499. [Google Scholar] [CrossRef]
  76. Kasetty, G.; Bhongir, R.K.V.; Papareddy, P.; Tufvesson, E.; Stenberg, H.; Bjermer, L.; Hultgårdh-Nilsson, A.; Herwald, H.; Egesten, A. Osteopontin protects against pneumococcal infection in a murine model of allergic airway inflammation. Allergy 2018, 74, 663–674. [Google Scholar] [CrossRef]
  77. Liu, W.; Zeng, Q.; Chen, Y.; Luo, R.Z. Role of Leptin/Osteopontin Axis in the Function of Eosinophils in Allergic Rhinitis with Obesity. Mediat. Inflamm. 2018, 2018, 9138904. [Google Scholar] [CrossRef]
  78. Goncalves DaSilva, A.; Liaw, L.; Yong, V.W. Cleavage of Osteopontin by Matrix Metalloproteinase-12 Modulates Experimental Autoimmune Encephalomyelitis Disease in C57BL/6 Mice. Am. J. Pathol. 2010, 177, 1448–1458. [Google Scholar] [CrossRef]
  79. Morimoto, Y.; Hirahara, K.; Kiuchi, M.; Wada, T.; Ichikawa, T.; Kanno, T.; Okano, M.; Kokubo, K.; Onodera, A.; Sakurai, D.; et al. Amphiregulin-Producing Pathogenic Memory T Helper 2 Cells Instruct Eosinophils to Secrete Osteopontin and Facilitate Airway Fibrosis. Immunity 2018, 49, 134–150. [Google Scholar] [CrossRef]
  80. Arjomandi, M.; Frelinger, J.; Donde, A.; Wong, H.; Yellamilli, A.; Raymond, W. Secreted Osteopontin Is Highly Polymerized in Human Airways and Fragmented in Asthmatic Airway Secretions. PLoS ONE 2011, 6, e25678. [Google Scholar] [CrossRef]
  81. Gela, A.; Bhongir, R.K.V.; Mori, M.; Keenan, P.; Mörgelin, M.; Erjefält, J.S.; Herwald, H.; Egesten, A.; Kasetty, G. Osteopontin That Is Elevated in the Airways during COPD Impairs the Antibacterial Activity of Common Innate Antibiotics. PLoS ONE 2016, 11, e0146192. [Google Scholar] [CrossRef]
  82. Papaporfyriou, A.; Loukides, S.; Kostikas, K.; Simoes, D.C.M.; Papatheodorou, G.; Konstantellou, E.; Hillas, G.; Papiris, S.; Koulouris, N.; Bakakos, P. Increased levels of osteopontin in sputum supernatant in patients with COPD. Chest 2014, 146, 951–958. [Google Scholar] [CrossRef]
  83. Lee, S.J.; Kim, S.H.; Kim, W.; Lim, S.; Lee, S.H.; Kim, Y.E.; Cho, Y.J.; Jeong, Y.Y.; Kim, H.C.; Lee, J.D.; et al. Increased plasma osteopontin in frequent exacerbator and acute exacerbation of COPD. Clin. Respir. J. 2014, 8, 305–311. [Google Scholar] [CrossRef]
  84. Hetman, O.; Krakhmalova, E.; Radzishevska, Y. The diagnostic value of osteopontin as an early marker of pilmonary hypertension affected by chronic obstructive pulmonary disease and concomitant ischemic heart disease. Georgian Med. News 2018, 41–447. [Google Scholar]
  85. Saker, M.; Lipskaia, L.; Marcos, E.; Abid, S.; Parpaleix, A.; Houssaini, A.; Validire, P.; Girard, P.; Noureddine, H.; Boyer, L.; et al. Osteopontin, a Key Mediator Expressed by Senescent Pulmonary Vascular Cells in Pulmonary Hypertension. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 1879–1890. [Google Scholar] [CrossRef] [Green Version]
  86. Maneechotesuwan, K.; Kasetsinsombat, K.; Wongkajornsilp, A.; Barnes, P.J. Simvastatin up-regulates adenosine deaminase and suppresses osteopontin expression in COPD patients through an IL-13-dependent mechanism. Respir. Res. 2016, 17, 104. [Google Scholar] [CrossRef]
  87. Dong, J.; Ma, Q. Myofibroblasts and lung fibrosis induced by carbon nanotube exposure. Part. Fibre Toxicol. 2016, 13, 60. [Google Scholar] [CrossRef]
  88. Dong, J.; Porter, D.W.; Batteli, L.A.; Wolfarth, M.G.; Richardson, D.L.; Ma, Q. Pathologic and molecular profiling of rapid-onset fibrosis and inflammation induced by multi-walled carbon nanotubes. Arch. Toxicol. 2015, 89, 621–633. [Google Scholar] [CrossRef]
  89. Mercer, R.R.; Scabilloni, J.F.; Hubbs, A.F.; Battelli, L.A.; McKinney, W.; Friend, S.; Wolfarth, M.G.; Andrew, M.; Castranova, V.; Porter, D.W. Distribution and fibrotic response following inhalation exposure to multi-walled carbon nanotubes. Part. Fibre Toxicol. 2013, 10, 33. [Google Scholar] [CrossRef]
  90. Dong, J.; Ma, Q. Osteopontin enhances multi-walled carbon nanotube-triggered lung fibrosis by promoting TGF-β1 activation and myofibroblast differentiation. Part. Fibre Toxicol. 2017, 14, 18. [Google Scholar] [CrossRef]
  91. Dadrich, M.; Nicolay, N.H.; Flechsig, P.; Bickelhaupt, S.; Hoeltgen, L.; Roeder, F.; Hauser, K.; Tietz, A.; Jenne, J.; Lopez, R.; et al. Combined inhibition of TGFβ and PDGF signaling attenuates radiation-induced pulmonary fibrosis. Oncoimmunology 2015, 5, e1123366. [Google Scholar] [CrossRef]
  92. Luo, Y.; Grötsch, B.; Hannemann, N.; Jimenez, M.; Ipseiz, N.; Uluckan, O.; Lin, N.; Schett, G.; Wagner, E.F.; Bozec, A. Fra-2 Expression in Osteoblasts Regulates Systemic Inflammation and Lung Injury through Osteopontin. Mol. Cell. Biol. 2018, 38, e00022-18. [Google Scholar] [CrossRef]
  93. Pardo, A.; Gibson, K.; Cisneros, J.; Richards, T.J.; Yang, Y.; Becerril, C.; Yousem, S.; Herrera, I.; Ruiz, V.; Selman, M.; et al. Up-regulation and profibrotic role of osteopontin in human idiopathic pulmonary fibrosis. PLoS Med. 2005, 2, e251. [Google Scholar] [CrossRef]
  94. White, E.S.; Xia, M.; Murray, S.; Dyal, R.; Flaherty, C.M.; Flaherty, K.R.; Moore, B.B.; Cheng, L.; Doyle, T.J.; Villalba, J.; et al. Plasma Surfactant Protein-D, Matrix Metalloproteinase-7, and Osteopontin Index Distinguishes Idiopathic Pulmonary Fibrosis from Other Idiopathic Interstitial Pneumonias. Am. J. Respir. Crit. Care Med. 2016, 194, 1242–1251. [Google Scholar] [CrossRef]
  95. Foster, M.W.; Morrison, L.D.; Todd, J.L.; Snyder, L.D.; Thompson, J.W.; Soderblom, E.J.; Plonk, K.; Weinhold, K.J.; Townsend, R.; Minnich, A.; et al. Quantitative proteomics of bronchoalveolar lavage fluid in idiopathic pulmonary fibrosis. J. Proteome Res. 2015, 14, 1238–1249. [Google Scholar] [CrossRef]
  96. Li, S.; Yang, R.; Sun, X.; Miao, S.; Lu, T.; Wang, Y.; Wo, Y.; Jiao, W. Identification of SPP1 as a promising biomarker to predict clinical outcome of lung adenocarcinoma individuals. Gene 2018, 679, 398–404. [Google Scholar] [CrossRef]
  97. He, F.; Ai, B.; Tian, L. Identification of genes and pathways in esophageal adenocarcinoma using bioinformatics analysis. Biomed. Rep. 2018, 9, 305–312. [Google Scholar] [CrossRef] [Green Version]
  98. Wisniewski, T.; Zyromska, A.; Makarewicz, R.; Zekanowska, E. Osteopontin And Angiogenic Factors as New Biomarkers of Prostate Cancer. Urol. J. 2018, 16, 134–140. [Google Scholar]
  99. Hauptman, N.; Boštjančič, E.; Žlajpah, M.; Ranković, B.; Zidar, N. Bioinformatics Analysis Reveals Most Prominent Gene Candidates to Distinguish Colorectal Adenoma from Adenocarcinoma. BioMed Res. Int. 2018, 2018, 9416515. [Google Scholar] [CrossRef]
  100. Tu, Y.; Fan, G.; Xi, H.; Zeng, T.; Sun, H.; Cai, X.; Kong, W. Identification of candidate aberrantly methylated and differentially expressed genes in thyroid cancer. J. Cell. Biochem. 2018, 119, 8797–8806. [Google Scholar] [CrossRef]
  101. Anborgh, P.H.; Lee, D.J.; Stam, P.F.; Tuck, A.B.; Chambers, A.F. Role of osteopontin as a predictive biomarker for anti-EGFR therapy in triple-negative breast cancer. Expert Opin. Ther. Targets 2018, 22, 727–734. [Google Scholar] [CrossRef]
  102. Treskova, I.; Topolcan, O.; Windrichova, J.; Simanek, V.; Slouka, D.; Treska, V.; Kucera, R. OPG, OPN, EGF and VEGF Levels at Individual Breslow Score Stages in Malignant Melanoma. Anticancer Res. 2018, 38, 4907–4911. [Google Scholar] [CrossRef]
  103. Ferreira, L.; Lima, R.; Bastos, A.; Silva, A.; Tavares, C.; Pestana, A.; Rios, E.; Eloy, C.; Sobrinho-Simões, M.; Gimba, E.; et al. OPNa Overexpression Is Associated with Matrix Calcification in Thyroid Cancer Cell Lines. Int. J. Mol. Sci. 2018, 19, 2990. [Google Scholar] [CrossRef]
  104. Hao, C.; Wang, Z.; Gu, Y.; Jiang, W.G.; Cheng, S. Prognostic Value of Osteopontin Splice Variant-c Expression in Breast Cancers: A Meta-Analysis. BioMed Res. Int. 2016, 2016, 7310694. [Google Scholar] [CrossRef]
  105. Cabiati, M.; Gaggini, M.; Cesare, M.M.; Caselli, C.; De Simone, P.; Filipponi, F.; Basta, G.; Gastaldelli, A.; Del Ry, S. Osteopontin in hepatocellular carcinoma: A possible biomarker for diagnosis and follow-up. Cytokine 2017, 99, 59–65. [Google Scholar] [CrossRef]
  106. Goparaju, C.M.V.; Pass, H.I.; Blasberg, J.D.; Hirsch, N.; Donington, J.S. Functional heterogeneity of osteopontin isoforms in non-small cell lung cancer. J. Thorac. Oncol. 2010, 5, 1516–1523. [Google Scholar] [CrossRef]
  107. Hao, C.; Cui, Y.; Hu, M.U.; Zhi, X.; Zhang, L.; Li, W.; Wu, W.; Cheng, S.; Jiang, W.G. OPN-a Splicing Variant Expression in Non-small Cell Lung Cancer and its Effects on the Bone Metastatic Abilities of Lung Cancer Cells In Vitro. Anticancer Res. 2017, 37, 2245–2254. [Google Scholar] [CrossRef] [Green Version]
  108. Zhao, B.; Sun, T.; Meng, F.; Qu, A.; Li, C.; Shen, H.; Jin, Y.; Li, W. Osteopontin as a potential biomarker of proliferation and invasiveness for lung cancer. J. Cancer Res. Clin. Oncol. 2011, 137, 1061–1070. [Google Scholar] [CrossRef]
  109. Hao, Y.; Liu, J.; Wang, P.; Wang, F.; Yu, Z.; Li, M.; Chen, S.; Ning, F. OPN Polymorphism Is Related to the Chemotherapy Response and Prognosis in Advanced NSCLC. Int. J. Genom. 2014, 2014, 846142. [Google Scholar] [CrossRef]
  110. Chen, Y.; Liu, H.; Wu, W.; Li, Y.; Li, J. Osteopontin genetic variants are associated with overall survival in advanced non-small-cell lung cancer patients and bone metastasis. J. Exp. Clin. Cancer Res. 2013, 32, 45. [Google Scholar] [CrossRef]
  111. Chang, Y.S.; Kim, H.J.; Chang, J.; Ahn, C.M.; Kim, S.K.; Kim, S.K. Elevated circulating level of osteopontin is associated with advanced disease state of non-small cell lung cancer. Lung Cancer 2007, 57, 373–380. [Google Scholar] [CrossRef]
  112. Sun, B.; Li, Y.; Zhang, Z.; You, J.; Wang, C. Osteopontin Combined with CD44v6, a Novel Prognostic Biomarker in Non-Small Cell Lung Cancer Undergoing Curative Resection. Ann. Thorac. Surg. 2013, 96, 1943–1951. [Google Scholar] [CrossRef]
  113. Chakraborty, G.; Jain, S.; Kundu, G.C. Osteopontin promotes vascular endothelial growth factor-dependent breast tumor growth and angiogenesis via autocrine and paracrine mechanisms. Cancer Res. 2008, 68, 152–161. [Google Scholar] [CrossRef]
  114. Liu, H.; Wei, S.; Zhang, L.; Yuan, C.; Duan, Y.; Wang, Q. Secreted Phosphoprotein 1 Promotes the Development of Small Cell Lung Cancer Cells by Inhibiting Autophagy and Apoptosis. Pathol. Oncol. Res. 2018. [Google Scholar] [CrossRef]
  115. Ouyang, X.; Huang, Y.; Jin, X.; Zhao, W.; Hu, T.; Wu, F.; Huang, J. Osteopontin promotes cancer cell drug resistance, invasion, and lactate production and is associated with poor outcome of patients with advanced non-small-cell lung cancer. Onco Targets Ther. 2018, 11, 5933–5941. [Google Scholar] [CrossRef]
  116. Saijo, A.; Goto, H.; Nakano, M.; Mitsuhashi, A.; Aono, Y.; Hanibuchi, M.; Ogawa, H.; Uehara, H.; Kondo, K.; Nishioka, Y. Bone marrow-derived fibrocytes promote stem cell-like properties of lung cancer cells. Cancer Lett. 2018, 421, 17–27. [Google Scholar] [CrossRef]
  117. Liu, F.; Bai, C.; Guo, Z. The prognostic value of osteopontin in limited-stage small cell lung cancer patients and its mechanism. Oncotarget 2017, 8, 70084–70096. [Google Scholar] [CrossRef] [Green Version]
  118. Choi, S.I.; Kim, S.Y.; Lee, J.H.; Kim, J.Y.; Cho, E.W.; Kim, I.-G. Osteopontin production by TM4SF4 signaling drives a positive feedback autocrine loop with the STAT3 pathway to maintain cancer stem cell-like properties in lung cancer cells. Oncotarget 2017, 8, 101284–101297. [Google Scholar] [CrossRef] [Green Version]
  119. Zhang, W.; Fan, J.; Chen, Q.; Lei, C.; Qiao, B.; Liu, Q. SPP1 and AGER as potential prognostic biomarkers for lung adenocarcinoma. Oncol. Lett. 2018, 15, 7028–7036. [Google Scholar] [CrossRef]
  120. Zhang, H.; Liu, H.; Yuan, D.; Wang, Z.; Wang, Y.; Song, Y. Prognostic value of secreted phosphoprotein-1 in pleural effusion associated with non-small cell lung cancer. BMC Cancer 2014, 14, 280. [Google Scholar] [CrossRef]
  121. Moschos, C.; Porfiridis, I.; Psallidas, I.; Kollintza, A.; Stathopoulos, G.T.; Papiris, S.A.; Roussos, C.; Kalomenidis, I. Osteopontin is upregulated in malignant and inflammatory pleural effusions. Respirology 2009, 14, 716–722. [Google Scholar] [CrossRef]
  122. Hsu, L.-H.; Hsu, P.-C.; Liao, T.-L.; Feng, A.-C.; Chu, N.-M.; Kao, S.-H. Pleural fluid osteopontin, vascular endothelial growth factor, and urokinase-type plasminogen activator levels as predictors of pleurodesis outcome and prognosticators in patients with malignant pleural effusion: A prospective cohort study. BMC Cancer 2016, 16, 463. [Google Scholar] [CrossRef]
  123. Giannou, A.D.; Marazioti, A.; Spella, M.; Kanellakis, N.I.; Apostolopoulou, H.; Psallidas, I.; Prijovich, Z.M.; Vreka, M.; Zazara, D.E.; Lilis, I.; et al. Mast cells mediate malignant pleural effusion formation. J. Clin. Investig. 2015, 125, 2317–2334. [Google Scholar] [CrossRef]
  124. Psallidas, I.; Stathopoulos, G.T.; Maniatis, N.A.; Magkouta, S.; Moschos, C.; Karabela, S.P.; Kollintza, A.; Simoes, D.C.M.; Kardara, M.; Vassiliou, S.; et al. Secreted phosphoprotein-1 directly provokes vascular leakage to foster malignant pleural effusion. Oncogene 2013, 32, 528–535. [Google Scholar] [CrossRef]
  125. Felten, M.K.; Khatab, K.; Knoll, L.; Schettgen, T.; Müller-Berndorff, H.; Kraus, T. Changes of mesothelin and osteopontin levels over time in formerly asbestos-exposed power industry workers. Int. Arch. Occup. Environ. Health 2014, 87, 195–204. [Google Scholar] [CrossRef]
  126. Bayram, M.; Dongel, I.; Akbaş, A.; Benli, İ.; Akkoyunlu, M.E.; Bakan, N.D. Serum Biomarkers in Patients with Mesothelioma and Pleural Plaques and Healthy Subjects Exposed to Naturally Occurring Asbestos. Lung 2014, 192, 197–203. [Google Scholar] [CrossRef]
  127. Bonotti, A.; Simonini, S.; Pantani, E.; Giusti, L.; Donadio, E.; Mazzoni, M.R.; Chella, A.; Marconi, L.; Ambrosino, N.; Lucchi, M.; et al. Serum mesothelin, osteopontin and vimentin: Useful markers for clinical monitoring of malignant pleural mesothelioma. Int. J. Biol. Markers 2017, 32, e126–e131. [Google Scholar] [CrossRef]
  128. Jamil, M.O.; Jerome, M.S.; Miley, D.; Selander, K.S.; Robert, F. A pilot study of zoledronic acid in the treatment of patients with advanced malignant pleural mesothelioma. Lung Cancer (Auckl) 2017, 8, 39–44. [Google Scholar] [CrossRef]
  129. Takeuchi, S.; Seike, M.; Noro, R.; Soeno, C.; Sugano, T.; Zou, F.; Uesaka, H.; Nishijima, N.; Matsumoto, M.; Minegishi, Y.; et al. Significance of osteopontin in the sensitivity of malignant pleural mesothelioma to pemetrexed. Int. J. Oncol. 2014, 44, 1886–1894. [Google Scholar] [CrossRef]
  130. Sun, B.; You, J.; Li, Y.; Zhang, Z.; Wang, C. Osteopontin knockdown suppresses non-small cell lung cancer cell invasion and metastasis. Chin. Med. J. 2013, 126, 1683–1688. [Google Scholar]
  131. Polat, B.; Wohlleben, G.; Katzer, A.; Djuzenova, C.S.; Technau, A.; Flentje, M. Influence of osteopontin silencing on survival and migration of lung cancer cells. Strahlenther. Onkol. 2013, 189, 62–67. [Google Scholar] [CrossRef]
  132. Ng, L.; Wan, T.M.-H.; Lam, C.S.-C.; Chow, A.K.-M.; Wong, S.K.-M.; Man, J.H.-W.; Li, H.-S.; Cheng, N.S.-M.; Pak, R.C.-H.; Cheung, A.H.-K.; et al. Post-operative plasma osteopontin predicts distant metastasis in human colorectal cancer. PLoS ONE 2015, 10, e0126219. [Google Scholar] [CrossRef]
  133. Kothari, A.N.; Arffa, M.L.; Chang, V.; Blackwell, R.H.; Syn, W.-K.; Zhang, J.; Mi, Z.; Kuo, P.C. Osteopontin-A Master Regulator of Epithelial-Mesenchymal Transition. J. Clin. Med. 2016, 5, 39. [Google Scholar] [CrossRef]
  134. Li, N.Y.; Weber, C.E.; Mi, Z.; Wai, P.Y.; Cuevas, B.D.; Kuo, P.C. Osteopontin up-regulates critical epithelial-mesenchymal transition transcription factors to induce an aggressive breast cancer phenotype. J. Am. Coll. Surg. 2013, 217, 17–26. [Google Scholar] [CrossRef]
  135. Raja, R.; Kale, S.; Thorat, D.; Soundararajan, G.; Lohite, K.; Mane, A.; Karnik, S.; Kundu, G.C. Hypoxia-driven osteopontin contributes to breast tumor growth through modulation of HIF1α-mediated VEGF-dependent angiogenesis. Oncogene 2014, 33, 2053–2064. [Google Scholar] [CrossRef]
  136. Dong, Q.; Zhu, X.; Dai, C.; Zhang, X.; Gao, X.; Wei, J.; Sheng, Y.; Zheng, Y.; Yu, J.; Xie, L.; et al. Osteopontin promotes epithelial-mesenchymal transition of hepatocellular carcinoma through regulating vimentin. Oncotarget 2016, 7, 12997–13012. [Google Scholar] [CrossRef]
  137. Jia, R.; Liang, Y.; Chen, R.; Liu, G.; Wang, H.; Tang, M.; Zhou, X.; Wang, H.; Yang, Y.; Wei, H.; et al. Osteopontin facilitates tumor metastasis by regulating epithelial–mesenchymal plasticity. Cell Death Dis. 2016, 7, e2564. [Google Scholar] [CrossRef]
  138. Shi, L.; Wang, X. Role of osteopontin in lung cancer evolution and heterogeneity. Semin. Cell Dev. Biol. 2017, 64, 40–47. [Google Scholar] [CrossRef]
  139. Giopanou, I.; Lilis, I.; Papaleonidopoulos, V.; Agalioti, T.; Kanellakis, N.I.; Spiropoulou, N.; Spella, M.; Stathopoulos, G.T. Tumor-derived osteopontin isoforms cooperate with TRP53 and CCL2 to promote lung metastasis. Oncoimmunology 2016, 6, e1256528. [Google Scholar] [CrossRef] [Green Version]
  140. Shao, L.; Zhang, B.; Wang, L.; Wu, L.; Kan, Q.; Fan, K. MMP-9-cleaved osteopontin isoform mediates tumor immune escape by inducing expansion of myeloid-derived suppressor cells. Biochem. Biophys. Res. Commun. 2017, 493, 1478–1484. [Google Scholar] [CrossRef]
  141. Kang, C.G.; Han, H.J.; Lee, H.-J.; Kim, S.-H.; Lee, E.-O. Rho-associated kinase signaling is required for osteopontin-induced cell invasion through inactivating cofilin in human non-small cell lung cancer cell lines. Bioorg. Med. Chem. Lett. 2015, 25, 1956–1960. [Google Scholar] [CrossRef]
  142. Li, Y.; Du, W.; Han, J.; Ge, J. LAMP3 promotes the invasion of osteosarcoma cells via SPP1 signaling. Mol. Med. Rep. 2017, 16, 5947–5953. [Google Scholar] [CrossRef]
  143. Kamioka, Y.; Takakura, K.; Sumiyama, K.; Matsuda, M. Intravital Förster resonance energy transfer imaging reveals osteopontin-mediated polymorphonuclear leukocyte activation by tumor cell emboli. Cancer Sci. 2017, 108, 226–235. [Google Scholar] [CrossRef]
  144. Turgut Cosan, D.; Oner, C.; Mutlu Sahin, F. Micro RNA-126 coordinates cell behavior and signaling cascades according to characteristics of breast cancer cells. Bratislavske lekarske listy 2016, 117, 639–647. [Google Scholar] [CrossRef]
  145. Coppola, D.; Szabo, M.; Boulware, D.; Muraca, P.; Alsarraj, M.; Chambers, A.F.; Yeatman, T.J. Correlation of osteopontin protein expression and pathological stage across a wide variety of tumor histologies. Clin. Cancer Res. 2004, 10, 184–190. [Google Scholar] [CrossRef]
  146. Rouanne, M.; Adam, J.; Goubar, A.; Robin, A.; Ohana, C.; Louvet, E.; Cormier, J.; Mercier, O.; Dorfmüller, P.; Fattal, S.; et al. Osteopontin and thrombospondin-1 play opposite roles in promoting tumor aggressiveness of primary resected non-small cell lung cancer. BMC Cancer 2016, 16, 483. [Google Scholar] [CrossRef]
  147. El-Tanani, M.K.; Yuen, H.-F.; Shi, Z.; Platt-Higgins, A.; Buckley, N.E.; Mullan, P.B.; Harkin, D.P.; Johnston, P.G.; Rudland, P.S. Osteopontin can act as an effector for a germline mutation of BRCA1 in malignant transformation of breast cancer-related cells. Cancer Sci. 2010, 101, 1354–1360. [Google Scholar] [CrossRef] [Green Version]
  148. Pass, H.I.; Lott, D.; Lonardo, F.; Harbut, M.; Liu, Z.; Tang, N.; Carbone, M.; Webb, C.; Wali, A. Asbestos exposure, pleural mesothelioma, and serum osteopontin levels. N. Engl. J. Med. 2005, 353, 1564–1573. [Google Scholar] [CrossRef]
  149. Paleari, L.; Rotolo, N.; Imperatori, A.; Puzone, R.; Sessa, F.; Franzi, F.; Meacci, E.; Camplese, P.; Cesario, A.; Paganuzzi, M. Osteopontin is not a specific marker in malignant pleural mesothelioma. Int. J. Biol. Markers 2009, 24, 112–117. [Google Scholar] [CrossRef]
  150. Hu, Z.-D.; Liu, X.-F.; Liu, X.-C.; Ding, C.-M.; Hu, C.-J. Diagnostic accuracy of osteopontin for malignant pleural mesothelioma: A systematic review and meta-analysis. Clin. Chim. Acta 2014, 433, 44–48. [Google Scholar] [CrossRef]
  151. Creaney, J.; Yeoman, D.; Musk, A.W.; de Klerk, N.; Skates, S.J.; Robinson, B.W.S. Plasma versus serum levels of osteopontin and mesothelin in patients with malignant mesothelioma--which is best? Lung Cancer 2011, 74, 55–60. [Google Scholar] [CrossRef]
  152. Cristaudo, A.; Foddis, R.; Bonotti, A.; Simonini, S.; Vivaldi, A.; Guglielmi, G.; Ambrosino, N.; Canessa, P.A.; Chella, A.; Lucchi, M.; et al. Comparison between plasma and serum osteopontin levels: Usefulness in diagnosis of epithelial malignant pleural mesothelioma. Int. J. Biol. Markers 2010, 25, 164–170. [Google Scholar] [CrossRef]
  153. Cappia, S.; Righi, L.; Mirabelli, D.; Ceppi, P.; Bacillo, E.; Ardissone, F.; Molinaro, L.; Scagliotti, G.V.; Papotti, M. Prognostic role of osteopontin expression in malignant pleural mesothelioma. Am. J. Clin. Pathol. 2008, 130, 58–64. [Google Scholar] [CrossRef]
  154. Rud, A.K.; Boye, K.; Oijordsbakken, M.; Lund-Iversen, M.; Halvorsen, A.R.; Solberg, S.K.; Berge, G.; Helland, A.; Brustugun, O.T.; Mælandsmo, G.M. Osteopontin is a prognostic biomarker in non-small cell lung cancer. BMC Cancer 2013, 13, 540. [Google Scholar] [CrossRef]
  155. Petta, V.; Loukides, S.; Kostikas, K.; Papaioannou, A.I.; Papatheodorou, G.; Cholidou, K.; Tomos, I.; Papiris, S.; Koulouris, N.G.; Bakakos, P. Serum osteopontin in patients with lung cancer and chronic obstructive pulmonary disease: Does the co-existence make the difference? J. Thorac. Dis. 2018, 10, 740–748. [Google Scholar] [CrossRef]
  156. Liaw, L.; Birk, D.E.; Ballas, C.B.; Whitsitt, J.S.; Davidson, J.M.; Hogan, B.L. Altered wound healing in mice lacking a functional osteopontin gene (spp1). J. Clin. Investig. 1998, 101, 1468–1478. [Google Scholar] [CrossRef]
  157. Berger, A.H.; Brooks, A.N.; Wu, X.; Shrestha, Y.; Chouinard, C.; Piccioni, F.; Bagul, M.; Kamburov, A.; Imielinski, M.; Hogstrom, L.; et al. High-throughput Phenotyping of Lung Cancer Somatic Mutations. Cancer Cell 2016, 30, 214–228. [Google Scholar] [CrossRef] [Green Version]
  158. Takafuji, V.; Forgues, M.; Unsworth, E.; Goldsmith, P.; Wang, X.W. An osteopontin fragment is essential for tumor cell invasion in hepatocellular carcinoma. Oncogene 2007, 26, 6361–6371. [Google Scholar] [CrossRef] [Green Version]
  159. Shen, H.; Weber, G.F. The osteopontin-c splice junction is important for anchorage-independent growth. Mol. Carcinog. 2014, 53, 480–487. [Google Scholar] [CrossRef]
  160. Zhang, G.; He, B.; Weber, G.F. Growth factor signaling induces metastasis genes in transformed cells: Molecular connection between Akt kinase and osteopontin in breast cancer. Mol. Cell. Biol. 2003, 23, 6507–6519. [Google Scholar] [CrossRef]
Figure 1. Structure of the human osteopontin (OPN) gene and protein variants. (A) Part of chromosome 4 with the SPP1 locus (red rectangle). (B) Structure of the SPP1 gene. White boxes, untranslated exons; black boxes, translated exons; black line, introns. (C) Exon composition of the protein variants resulting from alternative splicing.
Figure 1. Structure of the human osteopontin (OPN) gene and protein variants. (A) Part of chromosome 4 with the SPP1 locus (red rectangle). (B) Structure of the SPP1 gene. White boxes, untranslated exons; black boxes, translated exons; black line, introns. (C) Exon composition of the protein variants resulting from alternative splicing.
Cells 08 00815 g001
Figure 2. SPP1 expression in physiologic conditions and cancer. SPP1 mRNA expression was ranked as low, medium, and high. Low, <500 transcripts per million; medium, 500–1000 transcripts per million; high, >1000 transcripts per million. SPP1 protein expression was ranked as low, medium, and high. Low, expression by <25% of individuals tested; medium, expression by 25–50% of individuals tested; high, expression by >50% of individuals tested. RNA-seq and immunohistochemistry data were from The Human Protein Atlas (https://www.proteinatlas.org/).
Figure 2. SPP1 expression in physiologic conditions and cancer. SPP1 mRNA expression was ranked as low, medium, and high. Low, <500 transcripts per million; medium, 500–1000 transcripts per million; high, >1000 transcripts per million. SPP1 protein expression was ranked as low, medium, and high. Low, expression by <25% of individuals tested; medium, expression by 25–50% of individuals tested; high, expression by >50% of individuals tested. RNA-seq and immunohistochemistry data were from The Human Protein Atlas (https://www.proteinatlas.org/).
Cells 08 00815 g002
Figure 3. Overall survival in lung cancers according to SPP1 expression. Data and plot from http://kmplot.com/.
Figure 3. Overall survival in lung cancers according to SPP1 expression. Data and plot from http://kmplot.com/.
Cells 08 00815 g003
Table 1. Human OPN regulation. The bold font indicates the regulation process demonstrated in the lung.
Table 1. Human OPN regulation. The bold font indicates the regulation process demonstrated in the lung.
MoleculeActivationInhibition
Transcription factorsc-Myb [48]Smad4 [47]
ERG [49]
AML-1 [44]
C/EBPα [44]
SP-1 [45]
CBFA1 [50]
NF-κB [51]
Hormones1,25-dihydroxyvitamin D3 [52]
Incretins [53]
Inflammatory mediatorsINFγ 1 [54]INFγ 1 [52]
TNFα [54]INFβ [55]
IL-6 1 [54]PGE2 [56]
TGFβ [54]
IL-17a [54]
OtherInorganic phosphate [57]
VHL [46]
Glucose [53]
1 Antagonistic effects depending on cell type and disease.
Table 2. Overall survival in different cancers according to SPP1 expression. Data from http://kmplot.com/. NA indicates not applicable. ↘ and = indicate, respectively, worse and similar survival of patients with high SPP1 expression compared with patients with low SPP1 expression. The bold font represents thorax cancers.
Table 2. Overall survival in different cancers according to SPP1 expression. Data from http://kmplot.com/. NA indicates not applicable. ↘ and = indicate, respectively, worse and similar survival of patients with high SPP1 expression compared with patients with low SPP1 expression. The bold font represents thorax cancers.
Cancer TypePatient NumberCut-off Overall Survival in Months
Low SPP1 ExpressionHigh SPP1 ExpressionLog-rank P ValueSPP1 Effect on Overall Survival
Bladder 140592946305. 80 × 10−2
Breast 2395134852171631. 00 × 10−16
Cervical 1304788NANA9. 50 × 10−4
Esophageal 3161191145236. 30 × 10−2
Gastric876228931272. 90 × 10−1
Head and neck 1500501759311. 00 × 10−2
Kidney renal 153042,547118771. 10 × 10−1
Kidney renal papillary cell 3288140,81087501. 80 × 10−1
Liver364524284283. 50 × 10−6
Lung 219264151102575. 30 × 10−10
Lung adenocarcinoma7204305136751. 30 × 10−7
Lung squamous-cell carcinoma52414,78064333. 70 × 10−2
Ovarian1435896023187. 70 × 10−7
Pancreatic ductal 4177586673185. 90 × 10−4
Pheochromocytoma and Paraganglioma178223NANA4. 80 × 10−1=
Rectum 4165262052372. 10 × 10−2
Sarcoma25951687625. 40 × 10−2
Testicular Germ Cell134470NANA2. 20 × 10−1=
Thymoma119208NANA6. 70 × 10−2
Thyroid 3502308NANA3. 50 × 10−1=
Uterine 3405929104522. 20 × 10−1
1 squamous-cell carcinoma, 2 all histologies, 3 carcinoma, 4 adenocarcinoma.
Table 3. OPN expression vectors. Published plasmids from https://www.addgene.org/.
Table 3. OPN expression vectors. Published plasmids from https://www.addgene.org/.
OrganismMutationIsoformTagName
Human OPN-a pDONR223_SPP1_WT_V5 [157]
OPN-aGSTpGEX-6P1-OPNa-delta S [21]
OPN-a pCR3. 1-OPNa [21]
OPN-b pCR3. 1-OPNb [21]
OPN-bGSTpGEX-6P1-OPNb-delta S [21]
OPN-cGSTpGEX-6P1-OPNc-delta S [21]
OPN-c pCR3. 1-OPNc [21]
Base pairs 49–942OPN-aflagpDest490-OPN-a [158]
Base pairs 1–175, 217–942OPN-bflagpDest490-OPN-b [158]
EEKQ-->AAAAOPN-c pCR3. 1-OPNc M1 [159]
EEKQNAV-->AAAAAAAOPN-c pCR3. 1-OPNc M3 [159]
EEKQNA-->EEKNAOPN-c pCR3. 1-OPNc M4 [159]
EEKQNA-->EEKQANAOPN-c pCR3. 1-OPNc M5 [159]
SGSSEEKQNAVSSEET-->AGAAEEKQNAVAAEEAOPN-c pCR3. 1-OPNc PSM1 [159]
SGSSEEKQNAVSSEET-->AGAAEEKQNAVSSEETOPN-c pCR3. 1-OPNc PSM2 [159]
SGSSEEKQNAVSSEET-->SGSSEEKQNAVAAEEAOPN-c pCR3. 1-OPNc PSM3 [159]
SGSSEEKQNAVSSEET-->SGAAEEKQNAVSSEETOPN-c pCR3. 1-OPNc PSM4 [159]
SGSSEEKQNAVSSEET-->SGSSEEKQNAVAAEETOPN-c pCR3. 1-OPNc PSM5 [159]
Base pairs 1–93, 175–942OPN-c pDest490-OPN-c [158]
Base pairs 499–630 flagpDest490-OPN-10 kDa [158]
Base pairs 49–498 flagpDest490-OPN-NT [158]
Base pairs 631–942 flagpDest490-OPN-CT [158]
Mouse opn-1 mOPN-pcDNA [160]
opn-1 mOPN-PB0 [160]
opn-2EGFPpSpp1-is2 [123]
opn-3EGFPpSpp1-is3 [123]
antisense as-mOPN-PB0 [160]

Share and Cite

MDPI and ACS Style

Lamort, A.-S.; Giopanou, I.; Psallidas, I.; Stathopoulos, G.T. Osteopontin as a Link between Inflammation and Cancer: The Thorax in the Spotlight. Cells 2019, 8, 815. https://doi.org/10.3390/cells8080815

AMA Style

Lamort A-S, Giopanou I, Psallidas I, Stathopoulos GT. Osteopontin as a Link between Inflammation and Cancer: The Thorax in the Spotlight. Cells. 2019; 8(8):815. https://doi.org/10.3390/cells8080815

Chicago/Turabian Style

Lamort, Anne-Sophie, Ioanna Giopanou, Ioannis Psallidas, and Georgios T. Stathopoulos. 2019. "Osteopontin as a Link between Inflammation and Cancer: The Thorax in the Spotlight" Cells 8, no. 8: 815. https://doi.org/10.3390/cells8080815

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop