Full-length osteopontin and its splice variants as modulators of chemoresistance and radioresistance (Review)

  • Authors:
    • Etel R.P. Gimba
    • Mariana C.M. Brum
    • Gabriela Nestal De Moraes
  • View Affiliations

  • Published online on: December 6, 2018     https://doi.org/10.3892/ijo.2018.4656
  • Pages: 420-430
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Osteopontin (OPN) is a matricellular phosphoglycoprotein overexpressed in several tumor types and can activate several aspects of cancer progression in solid and non‑solid tumors. In the present review, the roles of OPN in mediating resistance to chemotherapy and radiotherapy and their main associated signaling pathways were summarized and discussed. Furthermore, it was detailed how OPN expression may be able to modulate resistance to these therapies by controlling epithelial cell plasticity, stemness potential and cell survival. Based on these data, the use of OPN and associated signaling was then proposed as potential molecular targets in order to sensitize resistant cells to main current therapeutic approaches. Finally, based on experimental evidence obtained by our group, the importance of investigating the specific roles OPN splicing isoforms have and how their properties may specifically control resistance to therapy was highlighted. These data elucidate a better understanding of how total OPN and their splicing isoforms, as well as their associated signaling, may contribute to main aspects of chemoresistance and radioresistance, such as those controlling cell survival, apoptosis, autophagy, stemness, epithelial cell plasticity and associated cell receptors.

1. Introduction

Cancer therapy resistance is mediated by several mechanisms, including intrinsic and extrinsic factors, and those originating from the tumor microenvironment (TME) (1). Most of the widely used chemotherapeutic agents and γ-radiation utilize apoptosis or autophagy as common death pathways. Thus, a better understanding of the molecular mechanisms behind tumor biology and cancer therapy resistance is a mandatory step to propose novel approaches aiming to bypass chemotherapy and radiotherapy resistance and associated gene products. Among several markers associated with response to therapy, osteopontin (OPN) has been identified as a key molecule (2,3).

OPN is a multi-functional chemokine-like matricellular phosphoglycoprotein. Depending on its intracellular or extracellular localization, OPN is involved in a series of physiological roles, including inflammation, cell adhesion and migration, differentiation, cell survival and apoptosis, as well as regulation of bone matrix mineralization. These diverse biological roles are partly due to its capacity to interact with several molecules, including cell surface receptors, such as integrin and cluster of differentiation (CD44), intracellular signaling molecules, calcium and heparin. OPN is produced by distinct cell types, such as epithelial, stromal, immune system, bone and endothelial cells (4). High OPN expression has also been detected in adipose tissues and body fluids (4).

In multiple cancer types, OPN expression is upregulated (3). In tumors, OPN regulates tumorigenesis, tumor progression and metastasis formation (5), by activating cell migration, inhibiting apoptosis (6,7), stimulating angiogenesis (8) and metabolism (9), and modulating the tumor microenvironment (10) and the immune system (11). Notably, OPN can also promote cell survival by negatively regulating apoptosis in response to stress conditions, including exposure to anticancer agents effect (12). OPN performs these roles by binding to cell surface receptors, including integrin and CD44 cell receptors (13).

Several aberrantly activated signaling pathways can activate OPN expression in cancer cells, such as activator protein-1, mycA, phosphatidylinositol 3 kinase (PI3K), serine/threonine kinase (AKT) and nuclear factor‑қB (NF‑қB) (14, 15). Notably, OPN has been described as a key modulator of cancer hallmarks, by regulating the main aspects of tumor progression in several tumor models (16,17). OPN is able to promote tumor cell migration by interacting with integrin receptors, especially αvβ3 integrin, stimulating cell adhesion and tumor cell migration properties (3). It has been demonstrated previously that OPN downregulation in breast cancer cell lines inhibits OPN interaction with αvβ3 integrin, thereby impairing cell migration, invasion and apoptosis (18). It was demonstrated that these effects have been mediated by PI3K/AKT/mechanistic target of rapamycin (mTOR) signaling, promoting the upregulation of light chain 3 and beclin-3, then favoring apoptotic cell death, while inhibiting aggressive phenotype of these breast cancer cells. It has also been demonstrated that alteration of OPN expression levels may influence tumor growth, migration and cell cycle in human nasopharyngeal CNE-2 carcinoma cell lines (18). When downregulating OPN expression, diminished levels of matrix metalloproteinase MMP-2 and MMP-9 have been observed, evidencing that OPN may also induce MMP expression levels through the activation of NF‑қB signaling in this tumor model (19). OPN expression levels can also modulate mitochondrial mediated apoptotic cell death, involving cytochrome c, apoptotic protease activating factor 1, cleaved caspase-3 and B cell lymphoma (Bcl)-2/Bcl-2-associated X protein, resulting in lower expression levels of proteins associated with cell invasion, such as MMP-2 and urokinase-type plasminogen activator (uPA) (20). Then, downregulation of OPN expression levels can promote apoptotic cell death and cell invasion properties in a mitochondrial-dependent pathway (20). OPN can also promote tumorigenesis and tumor progression by evading apoptotic cell death, mainly by interacting with CD44 cell receptors (21). Furthermore, OPN interactions with immune and inflammatory cells from the TME perform essential roles on tumor development and progression. Besides being produced by tumor cells, OPN can also be secreted by stromal and infiltrating inflammatory cells that can affect the TME and its corresponding cell roles. Stromal fibroblasts can also be influenced by OPN (22,23). Given its roles in angiogenesis, extracellular matrix remodeling and metastasis, their presence and influence by OPN can also favor tumor growth (22,23). It has been reported that when interacting with α9β1, OPN activates p38 and extracellular signal-regulated kinase (ERK) signaling, which then can promote the expression of cyclooxygenase-2 and prostaglandin E (PGE), favoring melanoma tumor cell migration (24). In addition, macrophages from the TME, when activated by OPN can promote angiogenesis via PGE2 and stimulate the expression of MMP-9, consequently promoting tumor progression in this tumor model (24). When recruiting macrophages to the tumor inflammatory environment, OPN can also stimulate tumorigenesis (25).

OPN also induces the expression and activity of MMPs, which can contribute to tumor metastasis by degrading the extracellular matrix (ECM) and promoting cell invasion. Conversely, OPN biological activity can also be modulated by MMP-induced cleavage (26). It is well-known that OPN stimulates tumor cell invasion and migration possibly by inhibiting apoptotic cell death and by regulating the activity of MMP-2 and MMP-9 that degrade the ECM (27). OPN can stimulate the activity of MMP-9, modulating multiple signaling pathways, such as focal adhesion kinase (FAK), ERK and NF‑қB, which then can regulate cytoskeleton architecture, cell growth, motility and extracellular matrix (ECM) (28,29). By activating PI3K/AKT signaling, OPN can similarly regulate hypoxia-inducible factor (HIF)-1α expression via αvβ3 integrin interaction and promoting ECM degradation through uPA and MMP-9, further mediating metastasis formation in ovarian cancer cells (30).

Also in the context of OPN roles on modulating the TME and the immune system, OPN has been described as a multifactorial cytokine activated by T lymphocytes, monocytes, macrophages, epithelial cells, fibroblasts and a promoter of cell-mediated immune responses (15,28).

Similarly, OPN is a typical angiogenesis stimulating factor, sustaining tumor progression and metastatic growth. The role of OPN in angiogenesis is mainly associated with OPN interaction with αvβ3 integrin, a central angiogenesis marker (31), but is also associated with several other factors, including vascular endothelial growth factor (VEGF) (32,33). OPN interactions with VEGF are also mediated by aberrant signaling pathways, such as PI3K/AKT and ERK1/ERK2 (32,33). It has also been demonstrated in acute leukemia that the expression of OPN and VEGF are strongly correlated with the occurrence and development of this non-solid tumor. In this model, OPN can regulate VEGF expression and promote angiogenesis besides favoring disease progression (31). It was also found that OPN interaction with αvβ3 activates signaling pathways, such as breast tumor kinase/NF‑қB/activating transcription factor (TF)-4, promoting cell migration, tumor growth, endothelial cell migration and angiogenesis (34). Our group also demonstrated that OPNb and OPNc splicing isoforms favored tumor growth, cell proliferation, invasion and migration by modulating VEGF, MMP-2 and MMP-9 expression levels through PI3K signaling (35).

It has also been demonstrated that OPN can modulate metabolism by signaling through the activation of oxidoreductase gene expression, associated with the mitochondrial respiratory chain, the hexose monophosphate shunt or the regulation of the hexose monophosphate shunt (9). Furthermore, it has been reported that OPN can disrupt liver cholesterol metabolism (36).

The majority of the data regarding the role of OPN in cancer cell refers to total OPN, which includes the sum of all OPN isoforms, including those generated by alternative splice, post‑translational modifications and alternative translation (17,37).

OPN primary transcript suffers alternative splicing, generating at least five splice variants, named OPNa (which contain all coding exons), OPNb (exon 5 is deleted), OPNc (exon 4 is deleted), OPN4 (in which both exon 4 and exon 5 are deleted) and OPN5 (which contains an additional exon originated from inclusion of a region from intron 3; Fig. 1). OPN splicing isoforms (OPN-SI) are aberrantly expressed in cancer cells (17). Particularly, the expression and functional roles of OPNa, OPNb and OPNc have been broadly studied in distinct tumor models, in which they were demonstrated to present tissue and tumor specific roles (17,37). Notably, the same OPN-SI might activate or inhibit tumor progression, depending on the tumor type (17,37). However, data regarding the expression and functional roles of OPN4 and OPN5 in tumor cells are scarce. One previous report described that these recently described OPN-SI are frequently co-expressed in esophageal carcinoma tissues (38), but functional studies evaluating these splice variants are still lacking.

In the present review, among the broadly known OPN roles on activating tumor progression, additional features involved in tumor resistance are highlighted. These typically include OPN functions on modulating cell survival, apoptosis and autophagy, as well as cell plasticity and sustaining stem-like properties of cancer cells (39), which are key factors associated with tumor resistance.

A molecular mechanism frequently associated with failure in the treatment of malignant carcinomas is the biological reprogramming of epithelial cells called epithelial mesenchymal plasticity. More recently, it has been defined as a dynamic process implicated in epithelial-mesenchymal transition (EMT) and its reverse program, mesenchymal-epithelial transition (MET) or intermediate phenotypes, known as partial or intermediate EMT (40,41). EMT renders cancer cells the ability to lose epithelial traits, while gaining mesenchymal features. During EMT, cells also acquire stem cell-like properties and are able to disseminate and colonize to distant organ sites, where they may exhibit elevated resistance to cancer therapies (42,43). Aberrant activation of oncogenic signaling pathways, including Wnt/β-catenin, hedgehog, Notch, PI3K-AKT, tumor necrosis factor-α and transforming growth factor (TGF)-β has a critical role in EMT (44) and occurs during acquisition of EMT phenotype and resistance to therapy. Furthermore, induction of resistance is also mediated by several genes regulated by the TF NF-κB, including Bcl-2, Bcl-xL, X-linked inhibitor of apoptosis (XIAP), survivin and AKT, which have been reported to mediate chemoresistance and radioresistance in numerous types of tumor cells (45).

In the present report, current knowledge regarding the OPN roles on mediating chemoresistance and radioresistance were reviewed, which are the primary cancer treatment approaches that are currently used. The distinct mechanisms by which OPN can promote resistance to specific drugs and their associated signaling pathways are also explored. Based on these data, putative treatment approaches targeting OPN that have been proposed to overcome resistance or inhibit tumor progression, and the particular contribution of OPN splice variants on the resistant phenotype are then discussed.

2. OPN and resistance to chemotherapy

OPN is able to mediate resistance to distinct chemotherapeutic drugs and in several cancer types. Table I summarizes these data and the corresponding signaling pathways and molecular mechanisms involved.

Table I

OPN roles in mediating chemoresistance and radioresistance.

Table I

OPN roles in mediating chemoresistance and radioresistance.

Author, yearDrug or therapyTumor typeBiological materialMain findingsSignaling pathways involved Refs.
Zhang et al, 2014PTLAMLAML cells resistant to DNROPN promotes resistance to PTL in CD34+/CD123+ leukemia stem cell population resistant to DNRPTL treatment downregulates OPN, and inhibits AKT, mTOR, β-catenin and PTEN(18)
Zhang et al, 2010SorafenibAMLTCGA data; MV4-11 human acute leukemia cell line (FLT3-ITD mutation)OPN binds to integrin αvβ3 and decrease sorafenib sensitivity in FLT3-ITD mutated AML cellsSorafenib sensitivity involves OPN/αvβ3 integrin/PI3K/ AKT/GSK3β/β-catenin pathway(20)
Gu et al, 2009CDDPHuman small cell lung cancerStable OPN overexpression in SBC-3 cells (SBC-3/OPN)Intracellular OPN mediates resistance to CDDP in SBC-3 cellsOPN induces anti-apoptotic Bcl-2 while blocking caspase-3 and -9(5)
Polyak et al, 2009CDDPOSCCSAS cellsOPN overexpression promotes resistance to CDDPNot investigated(23)
Luo et al, 20155-FUOSCCSAS cells overexpressing OPNOPN-αvβ3 integrin axis mediates resistance to 5-FUOPN-mediated resistance involves αvβ3 axis(50)
Ng et al, 2015OxaliplatinColorectal cancerDLD1 cells overexpressing OPNOPN expression and chemoresistance in patients treated with oxaliplatinOPN enhances stemness of cancer cells through CD44v6, HGF and SDF-1(51)
Lin et al, 2015Temozolomide and CDDPGliomaU251 human glioma cellsOPN mediates resistance to temozolomide and CDDPOPN-mediated resistance involves NF-κB/Bcl-2 pathway(52)
Shao et al, 2017CTXBreast cancerMDA-MB-231OPN knockdown sensitizes cells to CTXOPN-mediated resistance involves p38 MAPK pathway(26)
Guarneri et al, 2017Vinorelbin, etoposide and gemcitabineMalignant pleural mesotheliomaACC-MESO-1/OPN cells overexpressing OPNOPN is involved in multi drug resistance by enhancing the CD44 binding to HACD44 and HA binding and activation of PI3K signaling(27)
Wang et al, 2011RadiotherapyLung cancerNSCLC cell lines and xenograft tumorsOPN and EGFR pathway, have been associated with radiation resistance in lung cancer cellsRadiation resistance in tumors harboring KRAS mutations involves MLCC and CSC-like phenotypes(31)
Song et al, 2008RadiotherapyLung cancerA549 lung cancer cellsBeclin1-induced autophagy abrogates radioresistance by suppressing OPNRadioresistance is mediated by inhibition of Beclin1-induced autophagy(30)
Castello et al, 2017RadiotherapyCervical cancerRadiation resistant and sensitive LACSCC paraffin tissuesOPN and E-cadherin aberrant expression indicates radiation resistanceOPN-mediated EMT mediates radioresistance(32)

[i] OPN, osteopontin; 5-FU, fluorouracil; CDDP, cisplatin; PTL, parthenolide; OSCC, oral squamous cell carcinoma, Bcl-2, B-cell lymphoma-2; DNR, daunorubicin; AML, acute myeloid leukemia; PTEN, phosphatase and tensin homolog; HGF, hepatocyte growth factor; SDF-1, stromal cell-derived factor-1; CD, cluster of differentiation; CD44v6, variant isoform of CD44; CTX, cyclophosphamide; MLCC, mitosis-like condensed chromatin; LACSCC, locally advanced cervical squamous cell carcinoma; AKT, protein kinase B; mTOR, mechanistic target of rapamycin; TCGA, The Cancer Genome Atlas; GSK3β, glycogen synthase kinase 3β; NF-κB; nuclear factor-κB; MAPK, mitogen-activated protein kinase; HA, hyaluronan; PI3K, phosphoinositide 3-kinase; NSCLC, non-small cell lung carcinoma; EGFR, epidermal growth factor receptor; CSC, cancer stem cell; EMT, epithelial-mesenchymal transition.

In non-solid tumors, such as leukemia, OPN has been reported to mediate resistance to parthenolide and sorafenib. The natural compound parthenolide was demonstrated to induce apoptosis in daunorubicin-resistant stem-like leukemic cells through OPN downregulation and modulation of AKT, mTOR and β-catenin signaling (46). Particularly in acute myeloid leukemia, OPN was demonstrated to be upregulated in leukemic blasts, being correlated with a poor clinical outcome (47). Consistently, OPN promoted resistance to sorafenib by binding to αvβ3 integrin and by inducing β-catenin expression in an AKT and glycogen synthase kinase (GSK)3β-dependent manner (48).

Furthermore, there are several examples in which OPN mediates resistance in solid tumors, most of which focused on the roles of OPN in hepatocellular carcinomas (HCC). In these tumors, high OPN expression level is correlated with increased metastatic potential and resistance to taxanes or cisplatin. In lung cancer cells, intracellular OPN upregulation promotes cisplatin resistance by inducing the expression of anti-apoptotic protein Bcl-2 and by blocking caspase-3 and caspase-9 from activation (5). Also, OPN is able to stimulate HCC cell survival and autophagy, which then can favor stem cell-like properties and the resistant phenotype in response to epirubicin and cisplatin via binding with its receptor integrin αvβ3 and sustaining Forkhead box (Fox)O3a stability (42). It has been proposed that OPN may promote a cancer stem cell (CSC)-like phenotype via the αvβ3-NF-κB-HIF-1α pathway (49).

Other studies using human samples with oral squamous cell carcinoma demonstrated that OPN expression levels are correlated with therapy response and shorter overall survival. OPN upregulation in these tumors promoted resistance to cisplatin and to 5-fluorouracil and also involved the OPN-integrin αvβ3 axis (50). In addition, several reports described the association between upregulated OPN expression and chemoresistance in patients treated with oxaliplatin in colorectal cancer (51), cisplatin in lung cancer (5), temozolomide and cisplatin in glioma (52), cyclophosphamide in breast cancer (53) and vinorelbin, etoposide and gemcitabine in malignant pleural mesothelioma (54). Similarly to other tumor models, resistance to these chemotherapeutic drugs mainly involves the regulated expression of octamer-binding TF 4 and sex determining region Y-box (SOX)2 (modulators of stemness), Bcl-2, caspase-3 and caspase-9 (apoptosis regulators) (5,52), as well as NF-κB, AKT and p38/mitogen-activated protein kinase (MAPK) signaling (49,51). Furthermore, it has been demonstrated in malignant pleural mesothelioma that OPN could regulate chemosensitivity to vinorelbine, etoposide and gencitabin through the alteration of CD44 binding to hyaluronate (HA) (54). HA is a linear glycosaminoglycan that interacts with cell surface receptors, including CD44, facilitating cell adhesion, cell motility, cellular proliferation and tumor progression (54). OPN is strongly involved in multidrug resistance in this tumor by enhancing the CD44 binding to HA and PI3K/AKT signaling, thereby promoting cell survival and chemoresistance (54).

3. OPN and association to radioresistance

In addition to OPN roles in mediating chemoresistance, reports describing the functional association between OPN and radiotherapy resistance are scarce (Table I). However, a number of reports have described the roles of OPN as a marker of response to radiotherapy (55,56). OPN expression, in conjunction with the epidermal growth factor receptor pathway, have been associated with radiation resistance and poor prognosis in lung cancer, particularly in patients presenting tumors with KRAS mutations (57). It has also been reported in lung cancer that OPN is an indicator of resistance to radiotherapy. In a previous study, overexpression of beclin-1 induced cell death by autophagy in human lung cancer cells, reversing radioresistance (58). Radioresistance in these tumors has been associated with a stem cell-like phenotype and invasive potential (57). In cervical cancer, high OPN and low E-cadherin expression levels correlate with a radiation-resistant phenotype (59), further implicating OPN as a key molecule in the interface of radioresistance and cellular plasticity. Stem cell-like features and radioresistance in glioma cells can be promoted by OPN possibly via activation of CD44 signaling through its intracellular domain by enhancing HIF-2α activity (60). Fig. 2 summarizes the mechanisms and signaling pathways activated by OPN on promoting chemoresistance and radioresistance.

Figure 2

Cancer treatment resistance mechanisms mediated by OPN. Numerous and intricate mechanisms developed by cancer cells have been described at different levels in response to intracellular or extracellular OPN. Upon OPN binding to integrin receptors, especially by αV/β3 heterodimers, the PI3K/AKT/mTOR/β-catenin signaling pathway is triggered, leading to cell survival and chemotherapeutic resistance. OPN can also confer stem cell-like features to cancer cells by sustaining FoxO3a stability-induced autophagy as well as activating the αv/β3/NF-κB/HIF-1α pathway. Accordingly, OPN can modulate cancer stemness by altering CD44 receptor binding to hyaluronate and inducing AKT phosphorylation via CD44, thereby promoting cell survival and chemoresistance. In conjunction with the EGFR pathway, OPN expression, has been associated with radiation resistance particularly in tumors presenting KRAS mut, which was closely associated with the modulation of BIM and CRMP1 expression and thus, stem cell phenotype. Consistently, intracellular OPN was demonstrated to regulate the expression of Oct4 and Sox2, well known modulators of stemness. OPN expression levels have been demonstrated to negatively regulate p38 MAPK and correlate with caspase-3 and -9 blocking and expression of Bcl-2 anti-apoptotic protein, further contributing towards an apoptosis inactivation phenotype. Therefore, OPN has been demonstrated to modulate signaling pathways associated with chemoresistance and radioresistance, through promoting cell survival and inhibiting apoptosis as well as modulating autophagy and stemness in tumor cells. OPN, osteopontin; PI3K, phosphoinositide 3-kinase; AKT, protein kinase B; mTOR, mechanistic target of rapamycin; Fox, Forkhead box; NF-κB; nuclear factor-κB; HIF-1α, hypoxia-inducible factor-1α; Oct4, octamer-binding transcription factor 4; CD, cluster of differentiation; EGFR, epidermal growth factor receptor; mut, mutations; Bcl-2, B cell lymphoma-2; BIM, Bcl-2-like protein 11; CRMP1, collapsin response mediator protein 1; Sox2, sex determining region Y-box 2; MAPK, mitogen-activated protein kinase; GSK3β, glycogen synthase kinase 3β; CBP, cAMP response element binding protein binding protein.

4. OPN in the interface of epithelial plasticity and therapeutic resistance

EMT corresponds with the dynamic transdifferentiation of epithelial into mesenchymal cells, in which cells lose their epithelial features and become more motile mesenchymal cells (44). Increasing evidence has demonstrated that there is a close association between EMT and resistance to therapy (59,60), which may be caused by an enhancement of cancer cell survival, cell fate transition, and/or upregulation of drug resistance-related genes during the EMT transition.

The EMT process is mediated by several EMT-inducing TFs (EMT-TF), including TWIST1/2, SNAIL1/2 and zinc finger E-box binding homeobox (ZEB)1/2. In addition to these established EMT-TFs, other TFs have been demonstrated to induce or regulate EMT, including Fox TFs, GATA family, SOX, ovo-like transcriptional repressor 1/2 and grainyhead-like 2 (61,62). Mechanistically, EMT-TFs suppress the expression of key epithelial markers, such as E-cadherin, while activating the expression of mesenchymal genes, such as those coding for vimentin and fibronectin. Collectively, these regulatory networks control the integrity of and the balance between the epithelial and mesenchymal phenotypes. In chemoresistant cells, EMT-TFs increase cell survival in response to therapy-induced programmed cell-death by upregulating anti-apoptotic genes, while downregulating gene products performing pro-apoptotic roles (52). Certain EMT-TFs can also contribute to hormone therapy resistance by modulating the expression of their corresponding receptors (61). These findings make the EMT process an attractive target for reducing chemotherapy resistance (61).

It has also been demonstrated that EMT-TFs act cooperatively with changes at the RNA level that regulate EMT progression, such as alternative splicing and microRNA (miRNA or miR) and long non-coding RNA mediated control of EMT (61-64). EMT process is further controlled by multiple signaling pathways, in which multiple morphogenetic and environmental signals, such as TGF-β, WNT, epidermal and platelet-derived growth factors, inflammatory cytokines and integrin receptor ligands, have been demonstrated to promote EMT in response to extracellular signals (61). Among these molecules, NF-κB and matrix MMPs have also been identified as specific inducers of EMT. NF-κB has been described as a contributor to EMT, by combining with oncogenic gene products, such as RAS in order to protect cells from apoptosis (65). MMPs induce EMT associated with malignant transformation via a pathway dependent upon production of reactive oxygen species (ROS) and degrading matrix proteins, then favoring tumor cell migration and motility (65).

TGF-β, mostly TGF-β1 isoform, is the most well-studied cytokine in the induction of EMT. As reviewed elsewhere (61), TGF-β mediates EMT through mothers against decapentaplegic homolog (SMAD)-dependent or SMAD-independent pathways The SMAD-dependent pathway is initiated by binding of TGF-β1 to TGF-β receptor (TβR)II and TβRI, which then activate SMAD protein complex, which then enter the nucleus to induce the expression of lymphoid enhancer binding factor-1 TF. This TF binds to β-catenin, suppressing transcription of epithelial markers, while promoting the expression of mesenchymal markers. SMAD complexes not only activate the expression, but also increase the activity of EMT-TFs. Other changes in gene expression during EMT occur without directly requiring EMT-TFs, but are rather controlled by TGF-β-activated SMADs, which can directly activate the expression of certain mesenchymal genes. TGF-β also induces signaling through cytoplasmic expression of β-catenin, which is also modulated by the Wnt pathway. In addition to TGF-β-activated SMAD signals, TGF-β also induces EMT signaling through RHO-like GTPases, PI3K/AKT and MAPK pathways. Activation of RHO, RAC and cell division control protein 42 homolog GTPases drives actin reorganization, and lamellipodia and filopodia formation. PI3K signaling activation by TGF-β promotes the activation of mammalian TOR complex (mTORC1 and mTORC2). Notably, mTORC1 and mTORC2 modulate cell size, protein synthesis, motility and invasion. AKT decreases the level of SNAIL1 expression, attenuating E-cadherin repression and the activation of MMP-9 expression. AKT also phosphorylates GSK3β, resulting in SNAIL-1 stabilization. TGF-β also activates ERK, p38 and JUN N-terminal kinase/MAPK pathways, which then also increases TGF-β-induced transcription, leading to increased E-cadherin repression and activation of N-cadherin and MMP expression. EMT signaling has been demonstrated to induce the expression of genes coding for MMP-2 and MMP-9, which cleave Type IV collagen in the basal lamina to promote post-EMT invasion of underlying tissues. MMP-3 has also been demonstrated to directly induce EMT through activation of RAC1 GTPase-reactive oxygen species signaling, which promotes SNAIL1 expression. Additionally, SNAIL1/2 can also promote breakdown of the ECM via upregulation of MMPs (61). Furthermore, MMPs are capable of degrading E-cadherin in the cell membrane. All these processes enable cells to acquire a mesenchymal phenotype (61).

In this context, OPN has previously been reported as a master regulator of epithelial-mesenchymal plasticity, once it has an important regulatory role in the expression of key EMT regulators (66-71). OPN expression also shares functional interplay with the previously mentioned traditional EMT activators, such as TGF-β, TWIST 1/2, ZEB1/2 and SNAIL-family members. Importantly, OPN is able to guide EMT through specific cellular signaling pathways and by restructuring the TME to modify EMT processes (66-71).

OPN overexpression induces TWIST phosphorylation and/or activation through MAPK, AKT and/or RAC/AKT signaling pathways (71,72). Besides, OPN upregulates HIF-1α to induce EMT through TWIST activation and by maintaining the tumor cell stemness (66). TWIST also serves in OPN-mediated metastasis through activation of the PI3K/AKT pathway (66). Signaling mediated by OPN interacts directly or indirectly with ZEB TF family members, as well as leading to EMT. Notably, it is known that OPN is a potent activator of NF-κB (73), which induces the expression of both ZEB1/2 and therefore can regulate NF-κB/ZEB dependent EMT. Likewise, OPN can regulate ZEB-related EMT through non-NF-κB pathways, such as by upregulating the expression of miR-200 family members that inhibit ZEB1/2 initiated EMT (71). Otherwise, NF-κB has also been indicated as a key player of OPN and MMP-9 activation (74). Furthermore, OPN regulates EMT by overexpressing SNAIL EMT-TF. In addition, OPN co-regulates the expression of glioma-associated oncogene, a TF that mediates Sonic hedgehog signaling, which then induces the expression of SNAIL (71). Vimentin upregulation has also been induced by OPN (66). Notably, OPN induces the expression of TGF-β (71).

It has been reported that OPN is able to modify the tissue and TME to support EMT and hence can also indirectly modify EMT (66). During tumor progression, limited tumor oxygen availability and tumor metabolic demands induce hypoxia. As reviewed previously (60,61), the activities of HIF-1α and HIF-2α are enhanced in response to prolonged TGF-β and certain tyrosine kinase receptors. HIF-1α and HIF-2α can then activate EMT-TF expression and/or subcellular localization in tumor cells. Immune and stromal cells from the TME are also sources of EMT-inducing cytokine stimulation. It has been demonstrated that chemotherapy and radiotherapy promote oxidative and inflammatory stress in tumor tissues, contributing to increased expression of inflammatory cytokines and subsequent induction of the EMT program. In the TME, post-translational modifications of EMT-TF can also regulate the half-life of EMT-TFs (61). Malignant signals from the TME with long-lasting effects on associated cancer cells may also modulate epithelial plasticity and then sustain the metastatic potential and tumor chemoresistance. OPN modulates tumor-specific EMT by generating cancer-associated fibroblasts (CAFs), which secrete a multitude of factors in the TME that support tumor invasiveness and metastases, including TGF-β and interleukin-6. Tumor-derived OPN and exogenous OPN are able to induce transformation of CAFs from mesenchymal stem cells by stimulating the production of TGF-β. OPN also enhances the migration and invasion of malignant tumor cells through both the inhibition of apoptosis and by regulating the activities of MMP-2 and MMP-9, which degrade the ECM. OPN has been described to upregulate MMP-9 activity, modulating multiple signaling pathways via focal FAK, ERK and NF-κB that regulate cytoskeletal organization, cell motility, cell growth, and also cell migration, ECM invasion and tumor growth (61,62). In this context, it has been indicated that NF-κB may be a key player in OPN and MMP-9 activation (27). OPN is a substrate for several extracellular proteases and is cleaved in vitro and in vivo by MMPs. MMP-9, for instance, is known to cleave OPN (75), and it is also known that OPN-regulated signaling leads upregulation of MMP-2 in prostate cancer cells (74). Similarly, OPN can mediate the activation of MMP-9 during migration of prostate cancer and melanoma cells (74).

It has also been reported that OPN has an important role in regulating TGF-β-mediated processes and likely also regulates TGF-β-mediated EMT (70). Considering the emerging roles of OPN in modulating the EMT process, thus contributing to a drug resistant phenotype, the analysis of OPN expression has been considered as a potential target for future interventions to overcome tumor resistance. Notably, it has been reported that, unlike secreted OPN, which is able to trigger the EMT to initiate cancer metastasis, nuclear OPN is able to induce MET, contributing to metastasis establishment at secondary sites. In this model, OPN interacted with HIF-2α, latterly impacting on the AKT/mir-29/ZEB cascade (71). It has also demonstrated that VEGF in the TME is able to induce OPN nuclear translocation (71).

5. OPN as a target to overcome resistance to cancer therapy

Considering the emerging roles of OPN in several processes associated with chemoresistant and radioresistant phenotype, and its pleiotropic roles in the tumor cascade, the analysis of OPN expression and associated signaling has been considered as a potential target for future interventions trying to overcome tumor resistance, both to early and advanced tumors.

OPN-associated signaling pathways include PI3K/AKT, mTOR, β-catenin or phosphatase and tensin homolog (PTEN) gene expression (72), as well as αvβ3-NF-κB-HIF-1α (49), caspases and Bcl-2 (5,52), p38/MAPK (54) and GSK3β (48), among other targets. Advances in the understanding of the biology of tumor resistance, particularly the signaling pathways associated with this phenotype, may enable the development of novel approaches to overcome resistance to therapy.

Sp eci f ic a nd t a rgeted i n h ibit ion of t hese resistance-associated proteins and signaling pathways may potentially increase sensitivity of cancer cells to the cytotoxic action of chemotherapeutic agents and to radiation exposure (44,45). As an example of this approach to further sensitize cells to chemotherapy by targeting OPN and associated signaling, isolated primary CD34+/CD38- bone marrow derived acute myeloid leukemia (AML) cells have been treated with curcumin and daunorubicin in combination. This strategy induced AML cell growth inhibition and increased cytotoxicity by upregulating AKT, mTOR, β-catenin or PTEN. Notably, these effects were stronger when OPN expression was specifically knocked-down (72). It has also been reported that OPN/NF-κB-mediated autophagy is required for the maintenance of the stemness state of pancreatic cancer cells, which is associated with survival and chemoresistance (76). These data demonstrated that the blockade of autophagy by downregulating autophagy markers or by treating these pancreatic cells with an autophagy inhibitor reduced the pancreatic CSC populations and associated features, such as the expression of CD44, CD24, CD133 and aldehyde dehydrogenase 1 (76). Once OPN is able to stimulate autophagy and the expression of CSC markers (which includes integrin and CD44 receptors), these cell populations could be prevented by OPN downregulation approaches in order to sensitize pancreatic cancer cells to current chemotherapeutic drugs, such as gemcitabine (76).

Therapeutic application of small interfering RNA molecules targeting OPN (77,78) or neutralizing antibodies associated with OPN epitopes (79) have been tested in order to downregulate OPN expression levels and the results have been promising. An additional approach to sensitize cells to chemotherapy or radiotherapy would be using miRNA molecules targeting OPN or additional gene products associated with chemoresistance, such as those modulating EMT (80,81), drug transporters (78) and cell survival (81). Oncogenic miRNAs are the miRNAs with a defined role in cancer. Several miRNAs are deregulated in cancer cells and correlated with tumor features. Specifically, miRNAs have been reported to influence several tumor-related processes, such as EMT, tumor invasion, metastasis and resistance to therapy (80,82).

Among currently tested miRNAs targeting OPN, a number are able to modulate tumorigenicity, tumor growth and metastasis, such as miR-127-5p (83), hsa-miR-299-5p (80) and miR-181a (84). However, to the best of our knowledge, there is no report describing the specific effects of miRNAs on sensitizing tumor cells to chemotherapy. Conversely, a number of miRNAs that, via targeting OPN, may be promising tools to regulate chemoresistance and radioresistance mediated by OPN. It has been demonstrated that miR-127-5p and hsa-miR-299-5p are able to regulate OPN expression and can respectively modulate human chondrocyte cell proliferation and tumorigenicity, and also display vasculogenic mimicry of spheroid-forming breast cancer cells (80). Similarly, miR-181a regulation of OPN expression provides a novel mechanism of suppressing metastasis in cancer cell lines (84). Furthermore, three lentiviral vectors encoding miRNA against OPN have been reported to inhibit tumor growth and metastasis of human hepatocellular carcinoma, by decreasing MMP-2 and uPA expression, thus leading to inhibition of lung metastasis (85). Furthermore, RNA aptamers have also been proposed to target OPN and it has been demonstrated to decrease EMT and tumor growth (86,87).

Future studies aiming to improve the effects of chemotherapy and radiotherapy could propose similar strategies targeting OPN signaling and additional pathways associated with cancer cell survival and resistance.

6. OPN splice variants and their potential role in tumor resistance

Although previous studies have reported the expression and roles of OPN-SI regarding distinct aspects of tumor progression, data reporting the association between their expression and resistance to therapy are limited. A recent report proposed that OPNb and OPNc splicing isoforms are aberrantly expressed in leukemia cells in response to distinct chemotherapeutic drugs, such as daunorubicin, idarubicin and cytarabine, further mediating resistance to these drugs (88). However, it was not clearly demonstrated that specific knockdown of these splice variants reverted chemoresistance. Our group pioneered the studies of OPN-SI and their relation to chemoresistance, demonstrating that prostate cancer cells that ectopically overexpress OPNb or OPNc are more resistant to docetaxel (DXT) and display higher survival rates (89). The DXT-resistant phenotype was also associated with EMT features in which cells overexpressing OPNb or OPNc exhibited upregulated mesenchymal markers, as opposed to epithelial markers (89). In summary, these data demonstrated that OPN-SI differently modulate chemoresistance. As nuclear OPNc has been demonstrated as a prognostic marker in breast cancer (90) and also reported to be correlated with relapse (90,91) and poor survival (92), it is possible that nuclear OPNc may also be a potential marker of response to chemotherapy or radiotherapy, as has been previously reported (90). Future studies should further investigate the roles of OPN-SI in chemoresistance in distinct tumor models and also explore their involvement in radioresistance.

7. Conclusions

Growing evidence has pointed to the crucial role that OPN has in many aspects of cancer progression, including the acquisition of drug resistance. OPN not only induces integrin receptor-mediated oncogenic signaling pathways but also modulates the epithelial-mesenchymal phenotype and stemness, conferring cancer cells the ability to survive, proliferate, evade from cell death, migrate and colonize other tissues. Consequently, OPN-overexpressing cells are refractory to current treatment options as well as exhibit invasive and metastatic potential. Together, these findings provide evidence that OPN-triggered signaling pathways can be targeted to specifically induce cell death in chemo- and radio-insensitive cancer cells in order to overcome the therapeutic resistant phenotype. Future studies will uncover the role of specific OPN isoforms in the acquisition of treatment resistance and also address whether OPN and its isoforms may be reliable markers for cancer progression and poor response to standard therapy.

Funding

The present study has been funded by FAPERJ (grant nos. E-26/ 210.394/2014, E-26/010.002007/2014 and E-26/203.204/2015), CNPq (grant no. 310591/2014-7); Ministério da Sáude and UFF/Proppi (L'ORÉAL-UNESCO-ABC for Women in Science).

Availability of data and materials

Not applicable.

Authors' contributions

ERPG designed the manuscript, provided financial support, wrote the manuscript, prepared figures, edited and performed major revisions. MCB contributed to figure preparation and revision, as well as writing and editing the manuscript. GNM provided financial support, prepared figures, and contributed to writing and editing the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Gottesman MM, Lavi O, Hall MD and Gillet J-P: Toward a better understanding of the complexity of cancer drug resistance. Annu Rev Pharmacol Toxicol. 56:85–102. 2016. View Article : Google Scholar

2 

Bergman PJ and Harris D: Radioresistance, chemoresistance, and apoptosis resistance The past, present, and future. Vet Clin North Am Small Anim Pract. 27:47–57. 1997. View Article : Google Scholar : PubMed/NCBI

3 

Shevde LA and Samant RS: Role of osteopontin in the pathophysiology of cancer. Matrix Biol. 37:131–141. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Clemente N, Raineri D, Cappellano G, Boggio E, Favero F, Soluri MF, Dianzani C, Comi C, Dianzani U and Chiocchetti A: Osteopontin bridging innate and adaptive immunity in autoimmune diseases. J Immunol Res. 2016:76754372016. View Article : Google Scholar

5 

Gu T, Ohashi R, Cui R, Tajima K, Yoshioka M, Iwakami S, Sasaki S, Shinohara A, Matsukawa T, Kobayashi J, et al: Osteopontin is involved in the development of acquired chemo-resistance of cisplatin in small cell lung cancer. Lung Cancer. 66:176–183. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Huang RH, Quan YJ, Chen JH, Wang TF, Xu M, Ye M, Yuan H, Zhang CJ, Liu XJ and Min ZJ: Osteopontin promotes cell migration and invasion, and inhibits apoptosis and autophagy in colorectal cancer by activating the p38 MAPK signaling pathway. Cell Physiol Biochem. 41:1851–1864. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Wu Y, Jiang W, Wang Y, Wu J, Saiyin H, Qiao X, Mei X, Guo B, Fang X, Zhang L, et al: Breast cancer metastasis suppressor 1 regulates hepatocellular carcinoma cell apoptosis via suppressing osteopontin expression. PLoS One. 7:e429762012. View Article : Google Scholar : PubMed/NCBI

8 

Wu XL, Lin KJ, Bai AP, Wang WX, Meng XK, Su XL, Hou MX, Dong PD, Zhang JJ, Wang ZY, et al: Osteopontin knockdown suppresses the growth and angiogenesis of colon cancer cells. World J Gastroenterol. 20:10440–10448. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Shi Z, Wang B, Chihanga T, Kennedy MA and Weber GF: Energy metabolism during anchorage-independence Induction by osteopontin-c. PLoS One. 9:e1056752014. View Article : Google Scholar

10 

Malaponte G, Hafsi S, Polesel J, Castellano G, Spessotto P, Guarneri C, Canevari S, Signorelli SS, McCubrey JA and Libra M: Tumor microenvironment in diffuse large B-cell lymphoma: Matrixmetalloproteinases activation is mediated by osteopontin overexpression. Biochim Biophys Acta. 1863:483–489. 2016. View Article : Google Scholar

11 

Caputo S and Bellone M: Osteopontin and the immune system: Another brick in the wall. Cell Mol Immunol. 15:405–407. 2018. View Article : Google Scholar

12 

Mohammadi S, Ghaffari SH, Shaiegan M, Zarif MN, Nikbakht M, Akbari Birgani S, Alimoghadam K and Ghavamzadeh A: Acquired expression of osteopontin selectively promotes enrichment of leukemia stem cells through AKT/mTOR/PTEN/β-catenin pathways in AML cells. Life Sci. 152:190–198. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Bellahcène A, Castronovo V, Ogbureke KUE, Fisher LW and Fedarko NS: Small integrin-binding ligand N-linked glycoproteins (SIBLINGs): Multifunctional proteins in cancer. Nat Rev Cancer. 8:212–226. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Phillips RJ, Helbig KJ, Van der Hoek KH, Seth D and Beard MR: Osteopontin increases hepatocellular carcinoma cell growth in a CD44 dependant manner. World J Gastroenterol. 18:3389–3399. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Rabenstein M, Vay SU, Flitsch LJ, Fink GR, Schroeter M and Rueger MA: Osteopontin directly modulates cytokine expression of primary microglia and increases their survival. J Neuroimmunol. 299:130–138. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Cook AC, Tuck AB, McCarthy S, Turner JG, Irby RB, Bloom GC, Yeatman TJ and Chambers AF: Osteopontin induces multiple changes in gene expression that reflect the six ‘hallmarks of cancer’ in a model of breast cancer progression. Mol Carcinog. 43:225–236. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Gimba ER and Tilli TM: Human osteopontin splicing isoforms: Known roles, potential clinical applications and activated signaling pathways. Cancer Lett. 331:11–17. 2013. View Article : Google Scholar

18 

Zhang H, Guo M, Chen JH, Wang Z, Du XF, Liu PX and Li WH: Osteopontin knockdown inhibits αv,β3 integrin-induced cell migration and invasion and promotes apoptosis of breast cancer cells by inducing autophagy and inactivating the PI3K/Akt/mTOR pathway. Cell Physiol Biochem. 33:991–1002. 2014. View Article : Google Scholar

19 

Yang L, Wei L, Zhao W, Wang X, Zheng G, Zheng M, Song X and Zuo W: Down-regulation of osteopontin expression by RNA interference affects cell proliferation and chemotherapy sensitivity of breast cancer MDA-MB-231 cells. Mol Med Rep. 5:373–376. 2012.

20 

Zhang A, Liu Y, Shen Y, Xu Y and Li X: Osteopontin silencing by small interfering RNA induces apoptosis and suppresses invasion in human renal carcinoma Caki-1 cells. Med Oncol. 27:1179–1184. 2010. View Article : Google Scholar

21 

Naor D, Wallach-Dayan SB, Zahalka MA and Sionov RV: Involvement of CD44, a molecule with a thousand faces, in cancer dissemination. Semin Cancer Biol. 18:260–267. 2008. View Article : Google Scholar : PubMed/NCBI

22 

De Wever O, Demetter P, Mareel M and Bracke M: Stromal myofibroblasts are drivers of invasive cancer growth. Int J Cancer. 123:2229–2238. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Polyak K, Haviv I and Campbell IG: Co-evolution of tumor cells and their microenvironment. Trends Genet. 25:30–38. 2009. View Article : Google Scholar

24 

Kale S, Raja R, Thorat D, Soundararajan G, Patil TV and Kundu GC: Osteopontin signaling upregulates cyclooxygenase-2 expression in tumor-associated macrophages leading to enhanced angiogenesis and melanoma growth via α9β1 integrin. Oncogene. 33:2295–2306. 2014. View Article : Google Scholar

25 

Lin CN, Wang CJ, Chao YJ, Lai M-D and Shan Y-S: The significance of the co-existence of osteopontin and tumor-associated macrophages in gastric cancer progression. BMC Cancer. 15:1282015. View Article : Google Scholar : PubMed/NCBI

26 

Shao L, Zhang B, Wang L, Wu L, Kan Q and Fan K: MMP-9-cleaved osteopontin isoform mediates tumor immune escape by inducing expansion of myeloid-derived suppressor cells. Biochem Biophys Res Commun. 493:1478–1484. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Guarneri C, Bevelacqua V, Polesel J, Falzone L, Cannavò PS, Spandidos DA, Malaponte G and Libra M: NF-κB inhibition is associated with OPN/MMP 9 downregulation in cutaneous melanoma. Oncol Rep. 37:737–746. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Liu GX, Sun JT, Yang MX, Qi XM, Shao QQ, Xie Q, Qu X, Wei FC and Sun SZ: OPN promotes survival of activated T cells by up-regulating CD44 in patients with oral lichen planus. Clin Immunol. 138:291–298. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Matušan-Ilijaš K, Damante G, Fabbro D, Dorđević G, Hadžisejdić I, Grahovac M, Marić I, Spanjol J, Grahovac B, Jonjić N, et al: Osteopontin expression correlates with nuclear factor-κB activation and apoptosis downregulation in clear cell renal cell carcinoma. Pathol Res Pract. 207:104–110. 2011. View Article : Google Scholar

30 

Song G, Cai QF, Mao YB, Ming YL, Bao SD and Ouyang GL: Osteopontin promotes ovarian cancer progression and cell survival and increases HIF-1alpha expression through the PI3-K/Akt pathway. Cancer Sci. 99:1901–1907. 2008.PubMed/NCBI

31 

Wang HL, Ruan LH and Zhao XQ: Expression of osteopontin and VEGF in acute leukemia and their relationship with angiogenesis. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 19:926–929. 2011.In Chinese. PubMed/NCBI

32 

Castello LM, Raineri D, Salmi L, Clemente N, Vaschetto R, Quaglia M, Garzaro M, Gentilli S, Navalesi P, Cantaluppi V, et al: Osteopontin at the crossroads of inflammation and tumor progression. Mediators Inflamm. 2017:40490982017. View Article : Google Scholar : PubMed/NCBI

33 

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. 28:3412–3422. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Chakraborty G, Jain S and Kundu GC: Osteopontin promotes vascular endothelial growth factor-dependent breast tumor growth and angiogenesis via autocrine and paracrine mechanisms. Cancer Res. 68:152–161. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Tilli TM, Mello KD, Ferreira LB, Matos AR, Accioly MT, Faria PA, Bellahcène A, Castronovo V and Gimba ER: Both osteopontin-c and osteopontin-b splicing isoforms exert pro-tumorigenic roles in prostate cancer cells. Prostate. 72:1688–1699. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Nuñez-Garcia M, Gomez-Santos B, Buqué X, García-Rodriguez JL, Romero MR, Marin JJG, Arteta B, García-Monzón C, Castaño L, Syn WK, et al: Osteopontin regulates the cross-talk between phosphatidylcholine and cholesterol metabolism in mouse liver. J Lipid Res. 58:1903–1915. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Briones-Orta MA, Avendaño-Vázquez SE, Aparicio-Bautista DI, Coombes JD, Weber GF and Syn W-K: Osteopontin splice variants and polymorphisms in cancer progression and prognosis. Biochim Biophys Acta Rev Cancer. 1868:93–108. 108.A2017. View Article : Google Scholar : PubMed/NCBI

38 

Lin J, Myers AL, Wang Z, Nancarrow DJ, Ferrer-Torres D, Handlogten A, Leverenz K, Bao J, Thomas DG, Wang TD, et al: Osteopontin (OPN/SPP1) isoforms collectively enhance tumor cell invasion and dissemination in esophageal adenocarcinoma. Oncotarget. 6:22239–22257. 2015.PubMed/NCBI

39 

Choi SI, Kim SY, Lee JH, Kim JY, Cho EW and Kim IG: 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. 8:101284–101297. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Sui H, Zhu L, Deng W and Li Q: Epithelial-mesenchymal transition and drug resistance: Role, molecular mechanisms, and therapeutic strategies. Oncol Res Treat. 37:584–589. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Ye X and Weinberg RA: Epithelial-mesenchymal plasticity: A central regulator of cancer progression. Trends Cell Biol. 25:675–686. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Liu X and Fan D: The epithelial-mesenchymal transition and cancer stem cells: Functional and mechanistic links. Curr Pharm Des. 21:1279–1291. 2015. View Article : Google Scholar

43 

Li L and Li W: Epithelial-mesenchymal transition in human cancer: Comprehensive reprogramming of metabolism, epigenetics, and differentiation. Pharmacol Ther. 150:33–46. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Gonzalez DM and Medici D: Signaling mechanisms of the epithelial-mesenchymal transition. Sci Signal. 7:re82014. View Article : Google Scholar : PubMed/NCBI

45 

Li F and Sethi G: Targeting transcription factor NF-kappaB to overcome chemoresistance and radioresistance in cancer therapy. Biochim Biophys Acta. 1805:167–180. 2010.PubMed/NCBI

46 

Mohammadi S, Zahedpanah M, Ghaffari SH, Shaiegan M, Nikbakht M and Nikugoftar M: Osteopontin plays a unique role in resistance of CD34+/CD123+ human leukemia cell lines KG1a to parthenolide. Life Sci. 189:89–95. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Liersch R, Shin JW, Bayer M, Schwöppe C, Schliemann C, Berdel WE, Mesters R and Detmar M: Analysis of a novel highly metastatic melanoma cell line identifies osteopontin as a new lymphangiogenic factor. Int J Oncol. 41:1455–1463. 2012.PubMed/NCBI

48 

Yi H, Zeng D, Shen Z, Liao J, Wang X, Liu Y, Zhang X and Kong P: Integrin alphavbeta3 enhances β-catenin signaling in acute myeloid leukemia harboring Fms-like tyrosine kinase-3 internal tandem duplication mutations: Implications for micro-environment influence on sorafenib sensitivity. Oncotarget. 7:40387–40397. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Cao L, Fan X, Jing W, Liang Y, Chen R, Liu Y, Zhu M, Jia R, Wang H, Zhang X, et al: Osteopontin promotes a cancer stem cell-like phenotype in hepatocellular carcinoma cells via an integrin-NF-κB-HIF-1α pathway. Oncotarget. 6:6627–6640. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Luo SD, Chen YJ, Liu CT, Rau KM, Chen YC, Tsai HT, Chen CH and Chiu TJ: Osteopontin involves cisplatin resistance and poor prognosis in oral squamous cell carcinoma. BioMed Res Int. 2015:5085872015. View Article : Google Scholar : PubMed/NCBI

51 

Ng L, Wan T, Chow A, Iyer D, Man J, Chen G, Yau TC, Lo O, Foo CC, Poon JT, et al: Osteopontin overexpression induced tumor progression and chemoresistance to oxaliplatin through induction of stem-like properties in human colorectal cancer. Stem Cells Int. 2015:2478922015. View Article : Google Scholar : PubMed/NCBI

52 

Qian C, Li P, Yan W, Shi L, Zhang J, Wang Y, Liu H and You Y: Downregulation of osteopontin enhances the sensitivity of glioma U251 cells to temozolomide and cisplatin by targeting the NF-κB/Bcl 2 pathway. Mol Med Rep. 11:1951–1955. 2015. View Article : Google Scholar

53 

Pang H, Cai L, Yang Y, Chen X, Sui G and Zhao C: Knockdown of osteopontin chemosensitizes MDA-MB-231 cells to cyclophosphamide by enhancing apoptosis through activating p38 MAPK pathway. Cancer Biother Radiopharm. 26:165–173. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Tajima K, Ohashi R, Sekido Y, Hida T, Nara T, Hashimoto M, Iwakami S, Minakata K, Yae T, Takahashi F, et al: Osteopontin-mediated enhanced hyaluronan binding induces multidrug resistance in mesothelioma cells. Oncogene. 29:1941–1951. 2010. View Article : Google Scholar : PubMed/NCBI

55 

Wohlleben G, Scherzad A, Güttler A, Vordermark D, Kuger S, Flentje M and Polat B: Influence of hypoxia and irradiation on osteopontin expression in head and neck cancer and glioblastoma cell lines. Radiat Oncol. 10:1672015. View Article : Google Scholar : PubMed/NCBI

56 

Ostheimer C, Schweyer F, Reese T, Bache M and Vordermark D: The relationship between tumor volume changes and serial plasma osteopontin detection during radical radiotherapy of non-small-cell lung cancer. Oncol Lett. 12:3449–3456. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Wang M, Han J, Marcar L, Black J, Liu Q, Li X, Nagulapalli K, Sequist LV, Mak RH, Benes CH, et al: Radiation resistance in KRAS-mutated lung cancer is enabled by stem-like properties mediated by an osteopontin-EGFR pathway. Cancer Res. 77:2018–2028. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Chang SH, Minai-Tehrani A, Shin JY, Park S, Kim JE, Yu KN, Hong SH, Hong CM, Lee KH, Beck GR Jr, et al: Beclin1-induced autophagy abrogates radioresistance of lung cancer cells by suppressing osteopontin. J Radiat Res (Tokyo). 53:422–432. 2012. View Article : Google Scholar

59 

Huang X, Qian Y, Wu H, Xie X, Zhou Q, Wang Y, Kuang W, Shen L, Li K, Su J, et al: Aberrant expression of osteopontin and E-cadherin indicates radiation resistance and poor prognosis for patients with cervical carcinoma. J Histochem Cytochem. 63:88–98. 2015. View Article : Google Scholar :

60 

Pietras A, Katz AM, Ekström EJ, Wee B, Halliday JJ, Pitter KL, Werbeck JL, Amankulor NM, Huse JT and Holland EC: Osteopontin-CD44 signaling in the glioma perivascular niche enhances cancer stem cell phenotypes and promotes aggressive tumor growth. Cell Stem Cell. 14:357–369. 2014. View Article : Google Scholar : PubMed/NCBI

61 

van Staalduinen J, Baker D, Ten Dijke P and van Dam H: Epithelial-mesenchymal-transition-inducing transcription factors: New targets for tackling chemoresistance in cancer? Oncogene. Jul 12–2018.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

62 

Shibue T and Weinberg RA: EMT, CSCs, and drug resistance: The mechanistic link and clinical implications. Nat Rev Clin Oncol. 14:611–629. 2017. View Article : Google Scholar : PubMed/NCBI

63 

Vega S, Morales AV, Ocaña OH, Valdés F, Fabregat I and Nieto MA: Snail blocks the cell cycle and confers resistance to cell death. Genes Dev. 18:1131–1143. 2004. View Article : Google Scholar : PubMed/NCBI

64 

Lamouille S, Xu J and Derynck R: Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 15:178–196. 2014. View Article : Google Scholar : PubMed/NCBI

65 

Cichon MA and Radisky DC: ROS-induced epithelial-mesenchymal transition in mammary epithelial cells is mediated by NF-κB-dependent activation of Snail. Oncotarget. 5:2827–2838. 2014. View Article : Google Scholar : PubMed/NCBI

66 

Kothari AN, Arffa ML, Chang V, Blackwell RH, Syn WK, Zhang J, Mi Z and Kuo PC: Osteopontin-A Master Regulator of Epithelial-Mesenchymal Transition. J Clin Med. 5:52016. View Article : Google Scholar

67 

Yu X, Zheng Y, Zhu X, Gao X, Wang C, Sheng Y, Cheng W, Qin L, Ren N, Jia H, et al: Osteopontin promotes hepatocellular carcinoma progression via the PI3K/AKT/Twist signaling pathway. Oncol Lett. 16:5299–5308. 2018.PubMed/NCBI

68 

Li NY, Weber CE, Mi Z, Wai PY, Cuevas BD and Kuo PC: Osteopontin up-regulates critical epithelial-mesenchymal transition transcription factors to induce an aggressive breast cancer phenotype. J Am Coll Surg. 217:17–26. 2013. View Article : Google Scholar : PubMed/NCBI

69 

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. 7:12997–13012. 2016. View Article : Google Scholar : PubMed/NCBI

70 

Weber CE, Li NY, Wai PY and Kuo PC: Epithelial-mesenchymal transition, TGF-β, and osteopontin in wound healing and tissue remodeling after injury. J Burn Care Res. 33:311–318. 2012. View Article : Google Scholar : PubMed/NCBI

71 

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. 7:e25642016. View Article : Google Scholar : PubMed/NCBI

72 

Zahed Panah M, Nikbakht M, Sajjadi SM, Rostami S, Norooznezhad AH, Kamranzadeh Fumani H, Ghavamzadeh A and Mohammadi S: Anti-apoptotic effects of osteopontin via the up-regulation of AKT/mTOR/β-catenin loop in acute myeloid leukemia cells. Int J Hematol Oncol Stem Cell Res. 11:148–157. 2017.PubMed/NCBI

73 

Li X, Jiang Z, Li X and Zhang X: SIRT1 overexpression protects non-small cell lung cancer cells against osteopontin-induced epithelial-mesenchymal transition by suppressing NF-κB signaling. OncoTargets Ther. 11:1157–1171. 2018. View Article : Google Scholar

74 

Castellano G, Malaponte G, Mazzarino MC, Figini M, Marchese F, Gangemi P, Travali S, Stivala F, Canevari S and Libra M: Activation of the osteopontin/matrix metalloproteinase-9 pathway correlates with prostate cancer progression. Clin Cancer Res. 14:7470–7480. 2008. View Article : Google Scholar : PubMed/NCBI

75 

Tan TK, Zheng G, Hsu TT, Lee SR, Zhang J, Zhao Y, Tian X, Wang Y, Wang YM, Cao Q, et al: Matrix metalloproteinase-9 of tubular and macrophage origin contributes to the pathogenesis of renal fibrosis via macrophage recruitment through osteopontin cleavage. Lab Invest. 93:434–449. 2013. View Article : Google Scholar : PubMed/NCBI

76 

Yang MC, Wang HC, Hou YC, Tung HL, Chiu TJ and Shan YS: Blockade of autophagy reduces pancreatic cancer stem cell activity and potentiates the tumoricidal effect of gemcitabine. Mol Cancer. 14:1792015. View Article : Google Scholar : PubMed/NCBI

77 

Huang G, Du M-Y, Zhu H, Zhang N, Lu ZW, Qian LX, Zhang W, Tian X, He X and Yin L: MiRNA-34a reversed TGF-β-induced epithelial-mesenchymal transition via suppression of SMAD4 in NPC cells. Biomed Pharmacother. 106:217–224. 2018. View Article : Google Scholar : PubMed/NCBI

78 

Cui H, Zhang AJ, Chen M and Liu JJ: ABC transporter inhibitors in reversing multidrug resistance to chemotherapy. Curr Drug Targets. 16:1356–1371. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Zhang F, Luo W, Li Y, Gao S and Lei G: Role of osteopontin in rheumatoid arthritis. Rheumatol Int. 35:589–595. 2015. View Article : Google Scholar

80 

Shevde LA, Metge BJ, Mitra A, Xi Y, Ju J, King JA and Samant RS: Spheroid-forming subpopulation of breast cancer cells demonstrates vasculogenic mimicry via hsa-miR-299-5p regulated de novo expression of osteopontin. J Cell Mol Med. 14:1693–1706. 2010. View Article : Google Scholar

81 

Bhattacharya SD, Mi Z, Kim VM, Guo H, Talbot LJ and Kuo PC: Osteopontin regulates epithelial mesenchymal transition-associated growth of hepatocellular cancer in a mouse xenograft model. Ann Surg. 255:319–325. 2012. View Article : Google Scholar : PubMed/NCBI

82 

Wang J, Yang M, Li Y and Han B: The role of microRNAs in the chemoresistance of breast cancer. Drug Dev Res. 76:368–374. 2015. View Article : Google Scholar : PubMed/NCBI

83 

Liang J, Xu L, Zhou F, Liu AM, Ge HX, Chen YY and Tu M: MALAT1/miR-127-5p regulates osteopontin (OPN)-mediated proliferation of human chondrocytes through PI3K/Akt pathway. J Cell Biochem. 119:431–439. 2018. View Article : Google Scholar

84 

Boguslawska J, Sokol E, Rybicka B, Czubaty A, Rodzik K and Piekielko-Witkowska A: microRNAs target SRSF7 splicing factor to modulate the expression of osteopontin splice variants in renal cancer cells. Gene. 595:142–149. 2016. View Article : Google Scholar : PubMed/NCBI

85 

Sun BS, Dong QZ, Ye QH, Sun HJ, Jia HL, Zhu XQ, Liu DY, Chen J, Xue Q, Zhou HJ, et al: Lentiviral-mediated miRNA against osteopontin suppresses tumor growth and metastasis of human hepatocellular carcinoma. Hepatology. 48:1834–1842. 2008. View Article : Google Scholar : PubMed/NCBI

86 

Hunter C, Bond J, Kuo PC, Selim MA and Levinson H: The role of osteopontin and osteopontin aptamer (OPN-R3) in fibroblast activity. J Surg Res. 176:348–358. 2012. View Article : Google Scholar :

87 

Talbot LJ, Mi Z, Bhattacharya SD, Kim V, Guo H and Kuo PC: Pharmacokinetic characterization of an RNA aptamer against osteopontin and demonstration of in vivo efficacy in reversing growth of human breast cancer cells. Surgery. 150:224–230. 2011. View Article : Google Scholar : PubMed/NCBI

88 

Mirzaei A, Mohammadi S, Ghaffari SH, Nikbakht M, Bashash D, Alimoghaddam K and Ghavamzadeh A: Osteopontin b and c isoforms: Molecular Candidates Associated with Leukemic Stem Cell Chemoresistance in Acute Myeloid Leukemia. Asian Pac J Cancer Prev. 18:1707–1715. 2017.PubMed/NCBI

89 

Nakamura KDM, Tilli TM, Wanderley JL, Palumbo A Jr, Mattos RM, Ferreira AC, Klumb CE, Nasciutti LE and Gimba ER: Osteopontin splice variants expression is involved on docetaxel resistance in PC3 prostate cancer cells. Tumour Biol. 37:2655–2663. 2016. View Article : Google Scholar

90 

Zduniak K, Ziolkowski P, Ahlin C, Agrawal A, Agrawal S, Blomqvist C, Fjällskog ML and Weber GF: Nuclear osteopontin-c is a prognostic breast cancer marker. Br J Cancer. 112:729–738. 2015. View Article : Google Scholar : PubMed/NCBI

91 

Ortiz-Martínez F, Perez-Balaguer A, Ciprián D, Andrés L, Ponce J, Adrover E, Sánchez-Payá J, Aranda FI, Lerma E and Peiró G: Association of increased osteopontin and splice variant-c mRNA expression with HER2 and triple-negative/basal-like breast carcinomas subtypes and recurrence. Hum Pathol. 45:504–512. 2014. View Article : Google Scholar : PubMed/NCBI

92 

Patani N, Jiang W and Mokbel K: Osteopontin C mRNA expression is associated with a poor clinical outcome in human breast cancer. Int J Cancer. 122:26462008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2019
Volume 54 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gimba ER, Brum MC and Nestal De Moraes G: Full-length osteopontin and its splice variants as modulators of chemoresistance and radioresistance (Review). Int J Oncol 54: 420-430, 2019
APA
Gimba, E.R., Brum, M.C., & Nestal De Moraes, G. (2019). Full-length osteopontin and its splice variants as modulators of chemoresistance and radioresistance (Review). International Journal of Oncology, 54, 420-430. https://doi.org/10.3892/ijo.2018.4656
MLA
Gimba, E. R., Brum, M. C., Nestal De Moraes, G."Full-length osteopontin and its splice variants as modulators of chemoresistance and radioresistance (Review)". International Journal of Oncology 54.2 (2019): 420-430.
Chicago
Gimba, E. R., Brum, M. C., Nestal De Moraes, G."Full-length osteopontin and its splice variants as modulators of chemoresistance and radioresistance (Review)". International Journal of Oncology 54, no. 2 (2019): 420-430. https://doi.org/10.3892/ijo.2018.4656