Next Article in Journal
Single-Cell RNA-Seq Analysis Reveals Macrophages Are Involved in the Pathogenesis of Human Sporadic Acute Type A Aortic Dissection
Next Article in Special Issue
Stromal-Modulated Epithelial-to-Mesenchymal Transition in Cancer Cells
Previous Article in Journal
Imaging the Vulnerable Carotid Plaque with CT: Caveats to Consider. Comment on Wang et al. Identification Markers of Carotid Vulnerable Plaques: An Update. Biomolecules 2022, 12, 1192
Previous Article in Special Issue
Current Progress of EMT: A New Direction of Targeted Therapy for Colorectal Cancer with Invasion and Metastasis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Regulation of the Epithelial to Mesenchymal Transition in Osteosarcoma

1
Division of Orthopaedic Surgery, Department of Surgery, University of Alberta, Edmonton, AB T6G 2B7, Canada
2
Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R7, Canada
3
Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2R3, Canada
*
Author to whom correspondence should be addressed.
Biomolecules 2023, 13(2), 398; https://doi.org/10.3390/biom13020398
Submission received: 10 January 2023 / Revised: 9 February 2023 / Accepted: 14 February 2023 / Published: 20 February 2023
(This article belongs to the Special Issue EMT and Cancer II)

Abstract

:
The epithelial to mesenchymal transition (EMT) is a cellular process that has been linked to the promotion of aggressive cellular features in many cancer types. It is characterized by the loss of the epithelial cell phenotype and a shift to a more mesenchymal phenotype and is accompanied by an associated change in cell markers. EMT is highly complex and regulated via multiple signaling pathways. While the importance of EMT is classically described for carcinomas—cancers of epithelial origin—it has also been clearly demonstrated in non-epithelial cancers, including osteosarcoma (OS), a primary bone cancer predominantly affecting children and young adults. Recent studies examining EMT in OS have highlighted regulatory roles for multiple proteins, non-coding nucleic acids, and components of the tumor micro-environment. This review serves to summarize these experimental findings, identify key families of regulatory molecules, and identify potential therapeutic targets specific to the EMT process in OS.

1. Introduction

Osteosarcoma (OS) is a primary bone malignancy with an annual incidence of 2–4 per million [1]. It typically affects children, teens, and young adults [2], with a peak incidence from ages 10–19 [1], a second peak in adults over 60 [2], and a slight male preponderance [3]. The overall 5-year survival rate for OS is 60% but decreases to 27% in the presence of distant metastases [4]; the rate of metastases at diagnosis is 18% [5].
The origin of OS is poorly understood. As a sarcoma, it arises from mesenchymal cells, but it is not currently known whether the precursor cells are osteoblasts or mesenchymal stem cells [6]. Although the etiology of OS is largely a mystery, multiple risk factors have been identified. These include medical conditions such as hereditary retinoblastoma, Li-Fraumeni syndrome, Werner syndrome, Rothmund-Thompson syndrome, Bloom syndrome, and Paget’s disease [3]. Other risk factors include exposure to ionizing radiation and alkylating agents, both of which may have been used in the treatment of a prior malignancy [3].
The mainstay of treatment for osteosarcoma is surgical resection and frequently involves both neoadjuvant and adjuvant chemotherapy for higher grade tumors [7]. While advances in surgical techniques and chemotherapeutic regimens were associated with an initial improvement in outcomes, overall survival in OS has not significantly changed in several decades [8]. As medicine becomes more personalized, there is a growing interest in the identification of novel targeted therapies. A key component in developing targeted therapy is identifying specific pathways, proteins, or other molecules essential to cancer cell function. One of the cellular features often associated with aggressive cancers is the epithelial to mesenchymal transition (EMT).

2. EMT in Cancer

EMT is depicted in Figure 1. It is a process by which cells exhibiting an epithelial phenotype adopt a mesenchymal phenotype, which facilitates migration, invasion, and metastasis [9]. It exists in equilibrium with a reverse and complementary process, the mesenchymal to epithelial transition (MET), wherein cells revert back to an epithelial phenotype. Primary epithelial tumors exhibit epithelial cell markers such as E-cadherin. These cells demonstrate apical polarity, adhesion to a basement membrane, and tight cellular junctions [10]. For many cancers, EMT is critical in the early transition from normal to malignant cells. It is characterized by downregulation of epithelial cell markers, destabilization and loss of cell–cell junctions, loss of adherence to basement membrane and apical polarity, and cytoskeletal reorganization [9]. The result of these changes is a cell with mesenchymal morphology and characteristics.
Given the migratory potential of mesenchymal cells compared to epithelial cells, EMT has long been linked to cancer metastasis. However, inhibition of EMT has not been shown to affect the establishment of cancer metastases in vivo [11,12], and the cells found within metastatic tumors are more likely to exhibit an epithelial phenotype [12,13]. Despite this, tumor cells that have undergone EMT appear to drive local invasion and angiogenesis of the primary tumor [13]. These results suggest that EMT is critical for tumor invasion into the local vascular system, allowing cells to migrate to distant organs where secondary tumors are established largely by cells with an epithelial phenotype, which have a greater propensity for proliferation [9]. These may be either cells that have undergone EMT and subsequently MET or primary tumor cells that did not undergo EMT [13].
The molecular pathways associated with EMT are summarized in Figure 2. Zinc-finger E-box binding homeobox (ZEB), snail family transcriptional repressor 1 (SNAIL), snail family transcriptional repressor 2 (SLUG), and twist-related protein (TWIST) are well-known EMT transcription factors that are established downstream targets of multiple signaling pathways, including the canonical wnt/β-catenin pathway, the neurogenic locus notch homolog protein (Notch) pathway, the Transforming Growth Factor β/Suppressor of Mothers Against Decapentaplegic (TGFβ/SMAD) pathway, the phosphoinositide 3-kinase (PI3K)/Akt pathway, the mitogen-activated protein kinase (MAPK) pathway, the Ras/Raf/Mitogen-activated protein kinase/ERK kinase/extracellular-signal-regulated kinase (RAS/RAF/MEK/ERK) axis, and the Janus kinase-Signal Transducer and Activator of Transcription JAK/STAT pathway [10]. These signaling cascades often interact, share many intermediaries, and impact the regulation of one another. This presents a challenge for studying and targeting EMT, as the individual pathways are difficult to isolate.

3. EMT Signaling Pathways

3.1. TGFβ/SMAD Pathway

The TGFβ family of proteins includes three TGFβ isoforms, activins, and bone morphogenic proteins (BMPs) [14]. In EMT, TGFβs bind to TGFβ receptors (1/2), which initiate a signaling cascade, leading to the increased transcription of genes involved in EMT. Binding of TGFβ to its receptors (1/2) leads to phosphorylation of SMAD2 and SMAD3, which then form a complex with SMAD4. BMPs also bind TGFβ receptors, activating SMAD1 and SMAD5 and then forming a complex with SMAD4. These trimeric complexes migrate to the nucleus to act as transcription factors.
SMAD complexes activate the mesenchymal genes vimentin and fibronectin, as well as the EMT transcription factors Snail, Slug, Zinc finger E-box-binding homeobox 1 (ZEB1) and Twist. These, in turn, repress E-cadherin and can upregulate the expression of TGFβ ligands, establishing a positive feedback loop to maintain EMT [9,15,16].

3.2. Canonical wnt Pathway

The canonical wnt pathway is considered to be a key activator of EMT [9]. Signaling is initiated by a group of wnt ligands that bind to Frizzled receptors and trigger a cascade of events, leading to the nuclear translocation of β-catenin. β-catenin is constitutively produced in the cell and stored in cytosolic pools. In the absence of wnt signaling, phosphorylated β-catenin is associated with a destruction complex, ubiquitinated, and degraded by proteasomes. Following activation of the canonical wnt pathway, β-catenin is dephosphorylated and translocates to the nucleus, where it acts as a transcriptional co-factor to induce the expression of genes involved in cell differentiation, proliferation, and tumorigenesis [17,18].
This pathway has been directly implicated in EMT via the expression of Twist, Slug, N-cadherin, and the repression of E-cadherin [19]. The known EMT transcription factor Snail has been shown to positively regulate wnt signaling [20]. The inhibition of Secreted Frizzled Related Protein 1 (SFRP), a negative regulator of wnt ligands, has also been shown to have EMT-like effects in breast carcinoma cells in vitro, while sensitizing them to TGFβ-induced EMT [21].
β-catenin is also located at the cell membrane as part of an E-cadherin-containing multi-component adherens junction complex, which is a component of cell–cell interaction junctions. β-catenin contributes to anchoring E-cadherin, a transmembrane cell–cell adhesion protein at the cell surface to the intracellular actin cytoskeleton. β-catenin is released from the adherens complex upon disruption of these adherens junctions between cells. Once available in the cytosol, it enters the pathway described above and is either phosphorylated and degraded or, if the wnt pathway is active, dephosphorylated and translocated to the nucleus to function as a transcription factor for EMT-genes [22]. E-cadherin can therefore act as a negative regulator of the canonical wnt pathway by sequestering most of the β-catenin in the epithelial cell membrane.

3.3. Notch Pathway

The Notch pathway has been implicated in inducing EMT in both normal and neoplastic tissues, and is involved in controlling cell fate, differentiation, and proliferation. Four isoforms (Notch1 through Notch4) are known to bind Delta-like or Jagged family ligands. This interaction triggers a series of proteolytic events leading to the active fragment Notch Intracellular Domain (Notch ICD), which then acts in the nucleus, where it associates with binding partners and transcriptional activators [23]. Several components of the Notch pathway are highly expressed at the invasive margins of tumors, which express EMT markers such as vimentin, suggesting an important role for the Notch pathway in the regulation of EMT [24]. Notch acts via transcriptional regulation of ZEB, Snail, and Slug, which repress expression of E-cadherin and induce expression of vimentin and fibronectin [23,24,25].
There is crosstalk between the Notch and TGFβ pathway that occurs via SMADs. As described above, the SMAD family of proteins are integral to TGFβ signaling. They have also been shown to associate with Notch-ICD. This affects the expression of genes downstream of both Notch and TGFβ that are required for mesenchymal differentiation, a key component in EMT [26]. Silencing components of the Notch pathway have also been shown to prevent TGFβ-induced EMT [27].

3.4. Tyrosine Kinase Pathways

Mitogenic growth factors also play a role in the regulation of EMT. The binding of these growth factors causes their receptors to dimerize and induces the activation of both receptor and non-receptor tyrosine kinases (TKs). This enables the activation of several pathways—including the MAPK, JAK-STAT, and phosphatidylinositol 3-kinase-Akt (PI3-Akt) pathways. All of these have been implicated in EMT, and are involved in cell growth, proliferation, and migration [28]. PI3K/Akt has also been shown to play an important role in the regulation of transcriptionally active β-catenin, a key molecule in the previously discussed wnt signaling pathway [29]. Inhibition of TKs is a growing field of study in cancer therapeutics, with multiple inhibitors currently under investigation [30].
Inhibition of fibroblast growth factor (FGF), a mitogenic growth factor that participates in the induction of EMT via activation of the MAPK, induces the reverse process MET in vitro and delays tumor growth in vivo [31]. One isoform, FGF2, has been associated with reduced overall survival in several carcinoma types if overexpressed [31].
The binding of epidermal growth factor (EGF) to its receptor leads to activation of MAPK pathway and decreased expression of E-cadherin [32]. EGF also activates the JAK2-STAT3 pathway, which leads to EMT activation via Twist [33]. Additionally, EGF has been shown to induce EMT via TGFβ signaling and regulation of Snail [34] and phosphorylation of SMAD2/SMAD3 [35].
The activation of Akt, or Protein Kinase B, has been shown to upregulate the phosphorylation of Twist1 and inhibit apoptosis [36], and the inhibition of Akt has been shown to induce MET [37]. For example, hepatocyte growth factor (HGF) has been shown to activate EMT [38], which can be reversed via inhibition of the PI3K/Akt pathway. HGF was found to enhance tumor progression and metastasis of hepatocellular carcinoma in association with the c-MET receptor tyrosine kinase [39], a known activator of PI3K/Akt.

4. EMT in OS

As a mesenchymal cancer, the importance of EMT in OS has been disputed [40,41]. In fact, an early investigation including 4 clinical osteosarcoma samples by Sato et al. found there was no detectable E-cadherin expression in these cells [42], suggesting that the repression/downregulation of E-cadherin—a classically described step in EMT—would not be possible. In contrast, Yin et al. found that 20.6% of OS tissue samples expressed E-cadherin and those that did were less likely to metastasize, whereas the expression of Twist was significantly related to metastases and poorer overall survival [43]. The promotion of EMT in OS characterized by increased migration and invasion in vitro has been shown to be mediated via upregulation of Snail [44,45,46,47,48,49,50,51,52,53,54,55,56,57], Slug [58,59], Twist [60,61,62,63], and ZEB [64,65,66,67].
The following sections give an overview of studies that have examined the roles of different EMT regulatory molecules in OS. All of these were found to affect the expression of EMT-related cell-markers and are correlated closely with EMT-associated cellular features such as increased migration and invasion. Many also showed a link between their proposed EMT-regulatory molecule and OS metastases in vivo in animal models. Taken together, these results suggest that EMT does play a role in osteosarcoma and is associated with a more aggressive tumor phenotype. However, the term “transition” is not ideally suited to sarcoma cancers, and EMT may be better thought of as a set of pathways utilized to maintain and promote the existing mesenchymal phenotype.
Sannino et al. posited a possible hybrid phenotype in sarcoma tumors cells, utilizing the EMT and MET pathways to acquire both mesenchymal and epithelial characteristics that favor the initiation and establishment of distant metastases [40].
The highlighted pathways important in EMT regulation have all been shown to have a role in OS. TGFβs promote EMT and metastases in OS [68], and TGFβ inhibition has been shown to decrease EMT in OS [58,69,70,71,72,73,74]. Chen et al. also identified that estrogen-related receptor α (ERRα) upregulates TGF-β-mediated EMT in two OS cell lines [50]. Others have highlighted roles for MAPK [63,75,76,77] and JAK/STAT [52,78,79,80,81,82].
Notch signaling promotes proliferation, migration, and invasion of OS cells, and Notch overexpression increased tumor growth in vivo [83,84,85,86]. Notch inhibition reduced chemo-resistance in OS in vitro [87,88]. Wnt signaling has also been shown to mediate EMT in OS [49,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104]. It has been proposed that wnt signaling is particularly important in the pathogenesis of OS cancer stem cells [105].
TKs are of particular interest in OS, and multiple different TK proteins have been associated with aggressive cellular phenotypes. Many studies have demonstrated a regulatory role in OS for the downstream TK pathways PI3K/Akt [106,107,108,109,110,111,112,113,114] and RAS/RAF/MEK/ERK [61,115,116,117,118]. Multiple TK inhibitors have been a part of recently completed or ongoing clinical trials in the treatment of OS, including Apatinib, Axitinib, Cabozantinib, Cediranib, Crizotinib, Dasatinib, Imatinib, Pazopanib, Regorafenib, Sorafenib, and Sunitinib [119,120].

5. Regulation of EMT in OS—Proteins

As a complex and multi-faceted process, several proteins have been implicated in EMT regulation in OS [44,48,49,53,54,55,61,62,64,78,89,91,93,94,97,100,106,107,108,109,112,113,114,116,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151], and these are summarized in Table 1 and Table 2. These proteins were found to either promote [44,48,53,54,61,64,78,89,93,94,97,100,106,107,108,109,112,113,114,116,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146] or inhibit [49,55,62,91,147,148,149,150,151] EMT in vitro, and the majority were found to be correspondingly upregulated or downregulated in clinical OS tissue samples and/or established cell lines compared to normal controls. Each group of authors found a significant correlation between the studied protein and the levels of EMT-related proteins, such as E-cadherin, N-cadherin, and vimentin. They also reported a significant effect on aggressive cellular characteristics, such as migration and invasion ability in vitro. Where noted, the results were confirmed in vivo with mouse xenograft experiments.
A detailed review of the individual proteins investigated for their regulatory role in EMT of OS cells is outside of the scope of this review. Generally, their endogenous functions can be grouped into the following families: cell cycle regulation, immunity/inflammatory, cell signaling, cell structure, and metabolism. Each of these categories has a logical impact on EMT and/or cancer cell behavior.
Changes to cell differentiation and cell cycle regulation are recognized mechanisms by which normal cells can become cancerous. We identified 22 proteins with a regulatory role in EMT in OS whose endogenous functions impact these processes [53,54,62,64,78,91,97,100,112,116,126,130,131,139,140,141,142,143,144,145,146,151]. This group can be represented by several ubiquitin ligases [142,143] and deubiquitinases [97,145,146] that are known to target proteins critical for cell growth, proliferation, and differentiation. These were all found to be upregulated in clinical samples of osteosarcoma, and the overexpression or inhibition of these proteins was found to correlate with markers for EMT and OS cell proliferation.
The importance of immunity and inflammation on cancer progression is widely recognized [152], and these systems have also been implicated in the regulation of EMT [153]. Twelve of the EMT regulatory proteins described in Table 1 and Table 2 function as part of immunity and inflammation [44,48,107,109,114,123,124,127,132,135,137,150], many of which can be designated as “pro-inflammatory” proteins [107,114,123,124], and others that function as part of the development and activation of immune cells [44,48,109,127,137]. For example, the Programmed Death Ligand 2 (PD-L2) protein is a ligand for the Programmed Death 1 (PD-1) receptor, which is protective against T cell-mediated death in conjunction with tumor-associated macrophages [154]. Ren et al. found that PD-L2 knockdown decreased EMT and inhibited migration, invasion, and colony formation of OS cells in vitro, and reduced OS metastases in vivo in a mouse model [137].
Table 1. Effects of highly expressed proteins on EMT in OS.
Table 1. Effects of highly expressed proteins on EMT in OS.
ProteinIncreased Levels in Clinical SamplePromoted EMTPromoted Cell Migration/InvasionPromoted In Vivo Tumor GrowthPromoted In Vivo MetastasisEndogenous Function
ACTL6A [121]Yes Structure
AIM2 [114]No Immune
BMP-2 [93]No Cell Signaling
Calponin 3 [122]Yes Structure
Cathepsin E [123]Yes Immune
COPS3 [116]Yes ns Cell Cycle Regulation
COX2 [107]No Inflammation
CPE-ΔN [94]No Cell Signaling
cPLA2a [124]Yes Inflammation
CPXM2 [125]Yes Cell Signaling
Cul4A [126]Yes Inflammation
CXCR6 [127]Yes Immune
Cyr61 [61,128]No Cell Signaling
E2F1 [78]Yes Cell Cycle Regulation
EPB41L3 [129]Yes Structure
Fibulin-3 [89]Yes Structure
Fibulin-4 [106]Yes Structure
HOXB7 [130]Yes Cell Cycle Regulation
HuR [131]Yes Cell Cycle Regulation
ICSBP [44]Yes Immune
IL-33 [109]No Inflammation
MAGL [132]Yes Inflammation
Metadherin [133]No Cell Signaling
NETO2 [113]Yes Cell Signaling
OLR1 [134]Yes Cell Signaling
P2X7 [108]Yes Cell Signaling
PADI4 [135]Yes Inflammation
PDGFRβ [112]No Cell Cycle Regulation
PD-L2 [137]Yes Inflammation
PGI [136]No Metabolism
RIPK4 [100]Yes Cell Cycle Regulation
SenP1 [138]No Cell Signaling
SENP3 [139]Yes Cell Cycle Regulation
SIRT1 [64]Yes Cell Cycle Regulation
SOX3 [53]Yes Cell Cycle Regulation
SOX5 [54]Yes Cell Cycle Regulation
ST6Gal-1 [140]No Cell Cycle Regulation
TANK1 [141]No Cell Cycle Regulation
Tim-3 [48]Yes Immune
TRIM29 [142]Yes Cell Cycle Regulation
TRIM66 [143]Yes Cell Cycle Regulation
UHRF1 [144]No Cell Cycle Regulation
USP7 [97]Yes Cell Cycle Regulation
USP17 [145]Yes Cell Cycle Regulation
USP22 [146]Yes Cell Cycle Regulation
Biomolecules 13 00398 i001: Significant association; ns: No significant association; ↓: Inverse association; Biomolecules 13 00398 i002: Not studied/reported.
Table 2. Effects of poorly expressed proteins in OS.
Table 2. Effects of poorly expressed proteins in OS.
ProteinDecreased Levels in Clinical SampleInhibited EMTInhibited Cell Migration/InvasionInhibited In Vivo Tumor GrowthInhibited In Vivo MetastasisEndogenous Function
ARID1a [147]Yes Cell Signaling
Ezrin [148]No Structure
FTL [149]Yes Metabolism
GPER [55]Yes Cell Signaling
LAIR-1 [150]Yes Immune
RASSF4 [91]Yes Cell Cycle Regulation
SOX6 [62]Yes Cell Cycle Regulation
TSSC3 [49]Yes Cell Cycle Regulation
WWOX [151]Yes Cell Cycle Regulation
Biomolecules 13 00398 i003: Significant association; Biomolecules 13 00398 i004: Not studied/reported.
Many of the described proteins are implicated in cell signaling [55,93,94,108,113,126,129,134,135,139,148]. In addition to cell–cell interactions, this broad grouping includes the regulation of multiple cell processes that affect multiple other pathways and functions, including but not limited to cell cycle regulation, inflammation, immunity, and metabolism.
Finally, a subset of the proteins associated with EMT in OS are either structural proteins or regulate cell structure via interaction with the cytoskeleton [89,106,121,122,129,148]. This is perhaps the simplest and most logical grouping given the key morphological changes that take place during the EMT transformation, as depicted in Figure 1. Interestingly, Yuan et al. found that Erythrocyte Membrane Protein Band 4.1-like 3 (EPB41L3)—a cytoskeletal protein involved in cytoskeletal rearrangement, intracellular transport, and signal transduction—was increased in OS tissues and cell lines but was associated with an inhibition of EMT, migration, invasion, and cell viability in OS cell culture [129]. This pattern of expression was opposite to all of the other proteins impacting EMT in OS identified in this review.
When reported, the EMT pathways most implicated in these studies were wnt and PI3K/Akt. The nuclear localization and, therefore, transcriptional activity of the wnt/β-catenin pathway has also been shown to be regulated by PI3K [30], suggesting overlap in these EMT control mechanisms. The most frequently identified downstream target was Snail, which is known to promote EMT by suppressing E-cadherin expression [155], and further upregulates wnt signaling and EMT [20].

6. Regulation of EMT in OS—Non-Coding Ribonucleic Acids

Another key group of regulatory factors of EMT/MET in OS are non-coding ribonucleic acids (ncRNAs). These molecules have many forms and functions [156], one of which is gene regulation. Typically identified through queries to the Gene Expression Omnibus (GEO), the differential expression of multiple separate long non-coding RNAs (lncRNAs) [157], microRNAs (miRNAs) [158], circular RNAs (circRNAs) [159,160], and pseudogenes [161] have been found to relate to OS prognosis [162]. Table 3 and Table 4 summarize ncRNAs implicated in OS EMT regulation. Again, they were found to have a role in either promoting [57,63,66,76,85,86,90,95,98,99,101,103,109,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196] or inhibiting [47,49,51,65,77,92,96,102,104,111,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225] EMT and invasive cellular behaviors of OS cells in vitro, and there was significant overlap in the affected pathways and ultimate downstream targets. Very frequently there are multiple non-coding RNAs involved in the same pathway as they can also regulate other nucleic acids.
Unlike the pattern observed in the majority of these findings, Yuan et al. found that although erythrocyte membrane protein band 4.1-like 3 (EPB41L3) was upregulated in OS cell lines and clinical tissue samples, knockdown of EPB41L3 significantly increased the migration and invasion capacity of the investigated cell lines despite decreased cell viability [130]. The findings were similarly mixed for lncRNA NKILA [201] and miR-let-7d [225]. These studies highlight the complexity of EMT regulation in OS and suggest that it is only one possible factor relating to tumor behavior and prognosis.

7. Regulation of EMT in OS—The Tumor Microenvironment

There has been increased recognition of the importance of the tumor microenvironment on various cellular functions and characteristics. This is the three-dimensional structure surrounding tumor cells and comprises immune cells, vascular network, and extra-cellular matrix (ECM), among other components. The tumor microenvironment is unique not only for different cancer types but also for individual patients, and it is influenced by multiple factors, including patient sex and presence of metastases [226]. A better understanding of the interactions within the tumor micro-environment is expected to lead to the development of personalized treatments targeted at individual patients’ tumors.
Han et al. found that the presence of tumor-associated macrophages (TAMS) and the expression of the inflammatory marker cyclo-oxygenase 2 (COX2) correlated with OS metastases in clinical samples [80]. They also found co-culture of OS cells with TAMS promoted EMT and aggressive cellular features in vitro, which was reversible by COX2 inhibition. Additionally, COX2 inhibition reduced pulmonary metastases in vivo in a murine model [80]. Ling et al. found that Von Willebrand Factor (VWF)—which is secreted by the endothelial cells lining blood vessels—promoted EMT in vitro following OS and endothelial cell co-culture, as well as tumor growth and metastasis in vivo in a mouse model [227].
In addition to the cellular and biochemical makeup of the tumor microenvironment, the biomechanical properties of the ECM may also play a role in regulating EMT. Dai et al. developed a three-dimensional cell culture model with varying degrees of ECM stiffness [228]. This may be of particular relevance when evaluating OS tumors that exist in the bone—a relatively rigid environment—but eventually expand into the surrounding soft tissues, which are substantially less rigid. It may also account for some of the differences in OS metastatic patterns as more than 85% of metastatic OS occurs in the lungs, a soft tissue, compared to only 21% that occurs in the bone [229].

8. Targeting EMT in Osteosarcoma

Given its close association with aggressive and metastatic OS, EMT is a natural target for OS treatment. Treatments targeting EMT in OS include known medications, hormones, novel small molecules, and herbal medicines. The currently recommended chemotherapy regimen for OS includes doxorubicin, cisplatin, and high-dose methotrexate. At low doses, cisplatin has been shown to promote EMT, migration, invasion, and in vivo tumor growth [87]. However, these findings were reversible with inhibition of the EMT-related Notch pathway. EMT is recognized as an important factor contributing to chemotherapy resistance in multiple cancers [230], which was shown in OS by Ding et al., who found that OS cells induced to be resistant to methotrexate exhibited higher levels of EMT proteins and greater migration and invasion [231].
Other drugs that have not traditionally been used to treat osteosarcoma clinically but have been shown to inhibit EMT in OS in vitro include the cholesterol medication lovastatin [232] and zolendronate, a bisphosphonate medication used in the treatment of osteoporosis and other metabolic bone disorders [233,234]. In addition to the suppression of EMT, migration, and invasion, Kim et al. showed that OS cells and orthotopic tumors in mice had increased radiation-sensitivity following treatment with zolendronate, and this combination therapy was more effective than either treatment on its own [234].
Several hormone therapies have also been investigated for their effect on EMT in OS. These include estrogen, which inhibited EMT and promoted apoptosis of OS cells at high doses [235]. Treatment with irisin, a hormone derived from skeletal muscle, also suppressed OS EMT, cell proliferation, migration, and invasion [52]. Melatonin, a sleep-related hormone that is widely commercially available, has been shown to inhibit EMT [56,236,237] and OS cell migration and invasion in vitro, with additional results in vivo showing reduced metastasis in mice [237]. In contrast, treatment of OS cells with visfatin [45], a metabolic peptide first identified in visceral fat, induced EMT and increased cell migration and invasion. While these results do not suggest a direct role for visfatin in treatment of OS, further studies could examine the potential therapeutic effects of visfatin regulation.
Newer therapies with peptides and other small molecules allow for targeting more specific biologic functions, often associated with receptor inhibition. Inhibition of CXCR4 with Peptide R inhibited EMT, cell migration, and invasion in OS cells, and was thought to have the potential for less toxicity than existing CXCR4 inhibitors [238]. Similar suppression of EMT, inhibition of cell migration/invasion, and reduced tumorigenesis in vivo was observed with inhibition of vascular endothelial growth factor receptor-2 (VEGFR2) by Apatinib [239], Krüppel-like factor 5 (KFL5), and early growth response gene 1 (EGR1) by ML264 [81], and TGF-β by RepSox [70]. The 4′-aminochalcones D14 and D15 were found to inhibit EMT, cell migration, and invasion through upregulation of p53 [240].
The investigation of traditional and herbal medicines and their derivatives (both natural and synthetic) is a growing area of interest. The effect on EMT in OS has been studied for a number of these compounds [46,58,69,71,72,73,74,75,82,115,118,241,242,243,244,245,246,247,248,249], with results summarized in Table 5. While the majority of these inhibited both EMT and aggressive cellular characteristics, such as migration and invasion, Jiang et al. found that triptolide, a compound found in the vine Tripterygium wilfordii, increased EMT in OS cells in vitro but inhibited proliferation and invasion [249].
A non-pharmacological treatment targeting EMT in OS was also reported. Tumor-treating electrical field (TTEF) was reported to suppress EMT, cell migration, invasion, and angiogenesis of OS cells in culture via potential effects on VEGF and matrix metalloproteinase 2 (MMP2) [250].

9. Conclusions

EMT has significant implications in OS, despite its mesenchymal origin. Multiple studies have correlated changes in EMT with a more aggressive OS phenotype, both in vitro and in vivo. More than 100 proteins and non-coding nucleic acids have been identified as having a potential regulatory role in the OS EMT/MET pathways, and these may prove to be viable therapeutic targets and/or prognostic factors. These results should be interpreted with caution. While many of the studies discussed in this review confirmed the presence of their specific molecule of interest in clinical samples, most of the cell culture and animal studies were performed with only a handful of established cell lines. The majority of OS samples do not exhibit any E-cadherin and would therefore not experience a significant change secondary to E-cadherin suppression, a key process in EMT. It is possible that the cell lines most frequently utilized for these investigations are in the minority that do express E-cadherin and therefore exaggerate the EMT effect. Unfortunately, as OS is a rare cancer, any findings such as these are difficult to generalize. However, a role for EMT/MET has clearly been shown in cell culture and may well be a viable therapeutic target. Further work in additional cell lines or primary cell culture would help to confirm the findings outlined in this review.

Author Contributions

Conceptualization S.P. and K.H.; methodology, S.P., K.H. and A.K.; data curation, K.H. and A.K.; writing—original draft preparation K.H.; writing—review and editing, K.H., A.K., S.P. and P.P.; visualization, K.H.; supervision, S.P. and P.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rojas, G.A.; Hubbard, A.K.; Diessner, B.J.; Ribeiro, K.B.; Spector, L.G. International Trends in Incidence of Osteosarcoma (1988–2012). Int. J. Cancer 2021, 149, 1044–1053. [Google Scholar] [CrossRef]
  2. American Cancer Society Key Statistics for Osteosarcoma. Available online: https://www.cancer.org/cancer/osteosarcoma/about/key-statistics.html (accessed on 12 September 2021).
  3. Sadykova, L.R.; Ntekim, A.I.; Muyangwa-Semenova, M.; Rutland, C.S.; Jeyapalan, J.N.; Blatt, N.; Rizvanov, A.A. Epidemiology and Risk Factors of Osteosarcoma. Cancer Investig. 2020, 38, 259–269. [Google Scholar] [CrossRef]
  4. American Cancer Society Survival Rates for Osteosarcoma. Available online: https://www.cancer.org/cancer/osteosarcoma/detection-diagnosis-staging/survival-rates.html (accessed on 11 September 2021).
  5. Marko, T.A.; Diessner, B.J.; Spector, L.G. Prevalence of Metastasis at Diagnosis of Osteosarcoma: An International Comparison. Pediatr. Blood Cancer 2016, 63, 1006–1011. [Google Scholar] [CrossRef] [Green Version]
  6. de Azevedo, J.W.V.; de Medeiros Fernandes, T.A.A.; Fernandes, J.V.; de Azevedo, J.C.V.; Lanza, D.C.F.; Bezerra, C.M.; Andrade, V.S.; de Araújo, J.M.G.; Fernandes, J.V. Biology and Pathogenesis of Human Osteosarcoma (Review). Oncol. Lett. 2020, 19, 1099–1116. [Google Scholar] [CrossRef] [Green Version]
  7. Strauss, S.J.; Frezza, A.M.; Abecassis, N.; Bajpai, J.; Bauer, S.; Biagini, R.; Bielack, S.; Blay, J.Y.; Bolle, S.; Bonvalot, S.; et al. Bone Sarcomas: ESMO–EURACAN–GENTURIS–ERN PaedCan Clinical Practice Guideline for Diagnosis, Treatment and Follow-Up. Ann. Oncol. 2021, 32, 1520–1536. [Google Scholar] [CrossRef]
  8. Cole, S.; Gianferante, D.M.; Zhu, B.; Mirabello, L. Osteosarcoma: A Surveillance, Epidemiology, and End Results Program-Based Analysis from 1975 to 2017. Cancer 2022, 128, 2107–2118. [Google Scholar] [CrossRef]
  9. Brabletz, S.; Schuhwerk, H.; Brabletz, T.; Stemmler, M.P. Dynamic EMT: A Multi-tool for Tumor Progression. EMBO J. 2021, 40, e108647. [Google Scholar] [CrossRef]
  10. Dongre, A.; Weinberg, R.A. New Insights into the Mechanisms of Epithelial–Mesenchymal Transition and Implications for Cancer. Nat. Rev. Mol. Cell Biol. 2019, 20, 69–84. [Google Scholar] [CrossRef] [PubMed]
  11. Zheng, X.; Carstens, J.L.; Kim, J.; Scheible, M.; Kaye, J.; Sugimoto, H.; Wu, C.C.; Lebleu, V.S.; Kalluri, R. Epithelial-to-Mesenchymal Transition Is Dispensable for Metastasis but Induces Chemoresistance in Pancreatic Cancer. Nature 2015, 527, 525–530. [Google Scholar] [CrossRef] [Green Version]
  12. Fischer, K.R.; Durrans, A.; Lee, S.; Sheng, J.; Li, F.; Wong, S.T.C.; Choi, H.; el Rayes, T.; Ryu, S.; Troeger, J.; et al. Epithelial-to-Mesenchymal Transition Is Not Required for Lung Metastasis but Contributes to Chemoresistance. Nature 2015, 527, 472–476. [Google Scholar] [CrossRef] [Green Version]
  13. Lourenco, A.R.; Ban, Y.; Crowley, M.J.; Lee, S.B.; Ramchandani, D.; Du, W.; Elemento, O.; George, J.T.; Jolly, M.K.; Levine, H.; et al. Differential Contributions of Pre- And Post-EMT Tumor Cells in Breast Cancer Metastasis. Cancer Res. 2020, 80, 163–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Morikawa, M.; Derynck, R.; Miyazono, K. TGF-β and the TGF-β Family: Context-Dependent Roles in Cell and Tissue Physiology. Cold Spring Harb. Perspect. Biol. 2016, 8, a021873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Grande, M.T.; Sánchez-Laorden, B.; López-Blau, C.; de Frutos, C.A.; Boutet, A.; Arévalo, M.; Rowe, R.G.; Weiss, S.J.; López-Novoa, J.M.; Nieto, M.A. Snail1-Induced Partial Epithelial-to-Mesenchymal Transition Drives Renal Fibrosis in Mice and Can Be Targeted to Reverse Established Disease. Nat. Med. 2015, 21, 989–997. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Dhasarathy, A.; Phadke, D.; Mav, D.; Shah, R.R.; Wade, P.A. The Transcription Factors Snail and Slug Activate the Transforming Growth Factor-Beta Signaling Pathway in Breast Cancer. PLoS ONE 2011, 6, e26514. [Google Scholar] [CrossRef] [Green Version]
  17. Lamouille, S.; Xu, J.; Derynck, R. Molecular Mechanisms of Epithelial-Mesenchymal Transition. Nat. Rev. Mol. Cell Biol. 2014, 15, 178–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Clevers, H. Wnt/β-Catenin Signaling in Development and Disease. Cell 2006, 127, 469–480. [Google Scholar] [CrossRef] [Green Version]
  19. Wu, Y.; Ginther, C.; Kim, J.; Mosher, N.; Chung, S.; Slamon, D.; Vadgama, J. v Expression of Wnt3 Activates Wnt/β-Catenin Pathway and Promotes EMT-like Phenotype in Trastuzumab-Resistant HER2-Overexpressing Breast Cancer Cells. Mol. Cancer Res. 2012, 10, 1597–1606. [Google Scholar] [CrossRef] [Green Version]
  20. Stemmer, V.; de Craene, B.; Berx, G.; Behrens, J. Snail Promotes Wnt Target Gene Expression and Interacts with β-Catenin. Oncogene 2008, 27, 5075–5080. [Google Scholar] [CrossRef] [Green Version]
  21. Gauger, K.J.; Chenausky, K.L.; Murray, M.E.; Schneider, S.S. SFRP1 Reduction Results in an Increased Sensitivity to TGF-β Signaling. BMC Cancer 2011, 11, 59. [Google Scholar] [CrossRef] [Green Version]
  22. Balsamo, J.; Arregui, C.; Leung, T.C.; Lilien, J. The Nonreceptor Protein Tyrosine Phosphatase PTP1B Binds to the Cytoplasmic Domain of N-Cadherin and Regulates the Cadherin-Actin Linkage. J. Cell Biol. 1998, 143, 523–532. [Google Scholar] [CrossRef] [Green Version]
  23. Misiorek, J.O.; Przybyszewska-Podstawka, A.; Kałafut, J.; Paziewska, B.; Rolle, K.; Rivero-Müller, A.; Nees, M. Context Matters: Notch Signatures and Pathway in Cancer Progression and Metastasis. Cells 2021, 10, 94. [Google Scholar] [CrossRef] [PubMed]
  24. Natsuizaka, M.; Whelan, K.A.; Kagawa, S.; Tanaka, K.; Giroux, V.; Chandramouleeswaran, P.M.; Long, A.; Sahu, V.; Darling, D.S.; Que, J.; et al. Interplay between Notch1 and Notch3 Promotes EMT and Tumor Initiation in Squamous Cell Carcinoma. Nat. Commun. 2017, 8, 1758. [Google Scholar] [CrossRef] [Green Version]
  25. Timmerman, L.A.; Grego-Bessa, J.; Raya, A.; Bertrán, E.; Pérez-Pomares, J.M.; Díez, J.; Aranda, S.; Palomo, S.; McCormick, F.; Izpisúa-Belmonte, J.C.; et al. Notch Promotes Epithelial-Mesenchymal Transition during Cardiac Development and Oncogenic Transformation. Genes Dev. 2004, 18, 99–115. [Google Scholar] [CrossRef] [Green Version]
  26. Matsuno, Y.; Coelho, A.L.; Jarai, G.; Westwick, J.; Hogaboam, C.M. Notch Signaling Mediates TGF-Β1-Induced Epithelial-Mesenchymal Transition through the Induction of Snai1. Int. J. Biochem. Cell Biol. 2012, 44, 776–789. [Google Scholar] [CrossRef] [PubMed]
  27. Zavadil, J.; Cermak, L.; Soto-Nieves, N.; Böttinger, E.P. Integration of TGF-β/Smad and Jagged1/Notch Signalling in Epithelial-to-Mesenchymal Transition. EMBO J. 2004, 23, 1155–1165. [Google Scholar] [CrossRef] [Green Version]
  28. di Domenico, M.; Giordano, A. Signal Transduction Growth Factors: The Effective Governance of Transcription and Cellular Adhesion in Cancer Invasion. Oncotarget 2017, 8, 36869–36884. [Google Scholar] [CrossRef] [Green Version]
  29. Persad, A.; Venkateswaran, G.; Hao, L.; Garcia, M.E.; Yoon, J.; Sidhu, J.; Persad, S. Active β-Catenin Is Regulated by the PTEN/PI3 Kinase Pathway: A Role for Protein Phosphatase PP2A. Genes Cancer 2016, 7, 368–382. [Google Scholar] [CrossRef] [Green Version]
  30. Pottier, C.; Fresnais, M.; Gilon, M.; Jérusalem, G.; Longuespée, R.; Sounni, N.E. Tyrosine Kinase Inhibitors in Cancer: Breakthrough and Challenges of Targeted Therapy. Cancers 2020, 12, 731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Maehara, O.; Suda, G.; Natsuizaka, M.; Ohnishi, S.; Komatsu, Y.; Sato, F.; Nakai, M.; Sho, T.; Morikawa, K.; Ogawa, K.; et al. Fibroblast Growth Factor-2-Mediated FGFR/Erk Signaling Supports Maintenance of Cancer Stem-like Cells in Esophageal Squamous Cell Carcinoma. Carcinogenesis 2017, 38, 1073–1083. [Google Scholar] [CrossRef] [Green Version]
  32. Tashiro, E.; Henmi, S.; Odake, H.; Ino, S.; Imoto, M. Involvement of the MEK/ERK Pathway in EGF-Induced E-Cadherin down-Regulation. Biochem. Biophys. Res. Commun. 2016, 477, 801–806. [Google Scholar] [CrossRef]
  33. Lo, H.-W.; Hsu, S.-C.; Xia, W.; Cao, X.; Shih, J.-Y.; Wei, Y.; Abbruzzese, J.L.; Hortobagyi, G.N.; Hung, M.-C. Epidermal Growth Factor Receptor Cooperates with Signal Transducer and Activator of Transcription 3 to Induce Epithelial-Mesenchymal Transition in Cancer Cells via Up-Regulation of TWIST Gene Expression. Cancer Res. 2007, 67, 9066–9076. [Google Scholar] [CrossRef] [Green Version]
  34. Uttamsingh, S.; Bao, X.; Nguyen, K.T.; Bhanot, M.; Gong, J.; Chan, J.L.K.; Liu, F.; Chu, T.T.; Wang, L.H. Synergistic Effect between EGF and TGF-Β1 in Inducing Oncogenic Properties of Intestinal Epithelial Cells. Oncogene 2008, 27, 2626–2634. [Google Scholar] [CrossRef] [Green Version]
  35. Kim, J.; Kong, J.; Chang, H.; Kim, H.; Kim, A. EGF Induces Epithelial-Mesenchymal Transition through Phospho-Smad2/3-Snail Signaling Pathway in Breast Cancer Cells. Oncotarget 2016, 7, 85021–85032. [Google Scholar] [CrossRef] [Green Version]
  36. Vichalkovski, A.; Gresko, E.; Hess, D.; Restuccia, D.F.; Hemmings, B.A. PKB/AKT Phosphorylation of the Transcription Factor Twist-1 at Ser42 Inhibits P53 Activity in Response to DNA Damage. Oncogene 2010, 29, 3554–3565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Hong, K.O.; Kim, J.H.; Hong, J.S.; Yoon, H.J.; Lee, J.I.; Hong, S.P.; Hong, S.D. Inhibition of Akt Activity Induces the Mesenchymal-to-Epithelial Reverting Transition with Restoring E-Cadherin Expression in KB and KOSCC-25B Oral Squamous Cell Carcinoma Cells. J. Exp. Clin. Cancer Res. 2009, 28, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Grotegut, S.; von Schweinitz, D.; Christofori, G.; Lehembre, F. Hepatocyte Growth Factor Induces Cell Scattering through MAPK/Egr-1-Mediated Upregulation of Snail. EMBO J. 2006, 25, 3534–3545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Ogunwobi, O.O.; Puszyk, W.; Dong, H.J.; Liu, C. Epigenetic Upregulation of HGF and C-Met Drives Metastasis in Hepatocellular Carcinoma. PLoS ONE 2013, 8, e63765. [Google Scholar] [CrossRef] [PubMed]
  40. Sannino, G.; Marchetto, A.; Kirchner, T.; Grünewald, T.G.P. Epithelial-to-Mesenchymal and Mesenchymal-to-Epithelial Transition in Mesenchymal Tumors: A Paradox in Sarcomas? Cancer Res. 2017, 77, 4556–4561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Kahlert, U.D.; Joseph, J.V.; Kruyt, F.A.E. EMT- and MET-Related Processes in Nonepithelial Tumors: Importance for Disease Progression, Prognosis, and Therapeutic Opportunities. Mol. Oncol. 2017, 11, 860–877. [Google Scholar] [CrossRef] [Green Version]
  42. Sato, H.; Hasegawa, T.; Abe, Y.; Sakai, H.; Hirohashi, S. Expression of E-Cadherin in Bone and Soft Tissue Sarcomas: A Possible Role in Epithelial Differentiation. Hum. Pathol. 1999, 30, 1344–1349. [Google Scholar] [CrossRef]
  43. Yin, K.; Liao, Q.; He, H.; Zhong, D. Prognostic Value of Twist and E-Cadherin in Patients with Osteosarcoma. Med. Oncol. 2012, 29, 3449–3455. [Google Scholar] [CrossRef] [PubMed]
  44. Sung, J.Y.; Park, S.Y.; Kim, J.H.; Kang, H.G.; Yoon, J.H.; Na, Y.S.; Kim, Y.N.; Park, B.K. Interferon Consensus Sequence-Binding Protein (ICSBP) Promotes Epithelial-to-Mesenchymal Transition (EMT)-like Phenomena, Cell-Motility, and Invasion via TGF-β Signaling in U2OS Cells. Cell Death Dis. 2014, 5, e1224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Cheng, G.; Liu, C.; Sun, X.; Zhang, L.; Liu, L.; Ouyang, J.; Li, B. Visfatin Promotes Osteosarcoma Cell Migration and Invasion via Induction of Epithelial-Mesenchymal Transition. Oncol. Rep. 2015, 34, 987–994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Cheng, Z.; Guo, Y.; Yang, Y.; Kan, J.; Dai, S.; Helian, M.; Li, B.; Xu, J.; Liu, C. Nitidine Chloride Suppresses Epithelial-to-Mesenchymal Transition in Osteosarcoma Cell Migration and Invasion through Akt/GSK-3β/Snail Signaling Pathway. Oncol. Rep. 2016, 36, 1023–1029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Ru, N.; Liang, J.; Zhang, F.; Wu, W.; Wang, F.; Liu, X.; Du, Y. SPRY4 Intronic Transcript 1 Promotes Epithelial-Mesenchymal Transition Through Association with Snail1 in Osteosarcoma. DNA Cell Biol. 2016, 35, 290–295. [Google Scholar] [CrossRef]
  48. Feng, Z.M.; Guo, S.M. Tim-3 Facilitates Osteosarcoma Proliferation and Metastasis through the NF-ΚB Pathway and Epithelial-Mesenchymal Transition. Genet. Mol. Res. 2016, 15, gmr7844. [Google Scholar] [CrossRef] [PubMed]
  49. Lv, Y.F.; Dai, H.; Yan, G.; Meng, G.; Zhang, X.; Guo, Q. nan Downregulation of Tumor Suppressing STF CDNA 3 Promotes Epithelial-Mesenchymal Transition and Tumor Metastasis of Osteosarcoma by the Wnt/GSK-3β/β-Catenin/Snail Signaling Pathway. Cancer Lett. 2016, 373, 164–173. [Google Scholar] [CrossRef]
  50. Chen, Y.; Zhang, K.; Li, Y.; He, Q. Estrogen-Related Receptor α Participates Transforming Growth Factor-β (TGF-β) Induced Epithelial-Mesenchymal Transition of Osteosarcoma Cells. Cell Adh. Migr. 2017, 11, 338–346. [Google Scholar] [CrossRef] [Green Version]
  51. Zhang, Z.; Zhang, M.; Chen, Q.; Zhang, Q. Downregulation of MicroRNA-145 Promotes Epithelial-Mesenchymal Transition via Regulating Snail in Osteosarcoma. Cancer Gene Ther. 2017, 24, 83–88. [Google Scholar] [CrossRef]
  52. Kong, G.; Jiang, Y.; Sun, X.; Cao, Z.; Zhang, G.; Zhao, Z.; Zhao, Y.; Yu, Q.; Cheng, G. Irisin Reverses the IL-6 Induced Epithelial-Mesenchymal Transition in Osteosarcoma Cell Migration and Invasion through the STAT3/Snail Signaling Pathway. Oncol. Rep. 2017, 38, 2647–2656. [Google Scholar] [CrossRef] [Green Version]
  53. Qiu, M.; Chen, D.; Shen, C.; Shen, J.; Zhao, H.; He, Y. Sex-Determining Region Y-Box Protein 3 Induces Epithelial-Mesenchymal Transition in Osteosarcoma Cells via Transcriptional Activation of Snail1. J. Exp. Clin. Cancer Res. 2017, 36, 46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Zhang, D.; Liu, S. SOX5 Promotes Epithelial-Mesenchymal Transition in Osteosarcoma via Regulation of Snail. J. Balk. Union Oncol. 2017, 22, 258–264. [Google Scholar]
  55. Wang, Z.; Chen, X.; Zhao, Y.; Jin, Y.; Zheng, J. G-Protein-Coupled Estrogen Receptor Suppresses the Migration of Osteosarcoma Cells via Post-Translational Regulation of Snail. J. Cancer Res. Clin. Oncol. 2019, 145, 87–96. [Google Scholar] [CrossRef] [PubMed]
  56. Chen, Y.; Zhang, T.; Liu, X.; Li, Z.; Zhou, D.; Xu, W. Melatonin Suppresses Epithelial-to-Mesenchymal Transition in the MG-63 Cell Line. Mol. Med. Rep. 2020, 21, 1356–1364. [Google Scholar] [CrossRef]
  57. Wen, J.F.; Jiang, Y.Q.; Li, C.; Dai, X.K.; Wu, T.; Yin, W.Z. LncRNA-XIST Promotes the Oxidative Stress-Induced Migration, Invasion, and Epithelial-to-Mesenchymal Transition of Osteosarcoma Cancer Cells through MiR-153-SNAI1 Axis. Cell Biol. Int. 2020, 44, 1991–2001. [Google Scholar] [CrossRef]
  58. Sun, Y.; Jiang, X.; Lu, Y.; Zhu, J.; Yu, L.; Ma, B.; Zhang, Q. Oridonin Prevents Epithelial-Mesenchymal Transition and TGF-Β1-Induced Epithelial-Mesenchymal Transition by Inhibiting TGF-Β1/Smad2/3 in Osteosarcoma. Chem. Biol. Interact. 2018, 296, 57–64. [Google Scholar] [CrossRef]
  59. Sharili, A.S.; Allen, S.; Smith, K.; Price, J.; McGonnell, I.M. Snail2 Promotes Osteosarcoma Cell Motility through Remodelling of the Actin Cytoskeleton and Regulates Tumor Development. Cancer Lett. 2013, 333, 170–179. [Google Scholar] [CrossRef] [Green Version]
  60. Ishikawa, T.; Shimizu, T.; Ueki, A.; Yamaguchi, S.I.; Onishi, N.; Sugihara, E.; Kuninaka, S.; Miyamoto, T.; Morioka, H.; Nakayama, R.; et al. Twist2 Functions as a Tumor Suppressor in Murine Osteosarcoma Cells. Cancer Sci. 2013, 104, 880–888. [Google Scholar] [CrossRef]
  61. Hou, C.H.; Lin, F.L.; Hou, S.M.; Liu, J.F. Cyr61 Promotes Epithelial-Mesenchymal Transition and Tumor Metastasis of Osteosarcoma by Raf-1/MEK/ERK/Elk-1/TWIST-1 Signaling Pathway. Mol. Cancer 2014, 13, 236. [Google Scholar] [CrossRef] [Green Version]
  62. Wang, Z.; Li, J.; Li, K.; Xu, J. SOX6 Is Downregulated in Osteosarcoma and Suppresses the Migration, Invasion and Epithelial-Mesenchymal Transition via TWIST1 Regulation. Mol. Med. Rep. 2018, 17, 6803–6811. [Google Scholar] [CrossRef]
  63. Shi, D.; Wu, F.; Mu, S.; Hu, B.; Zhong, B.; Gao, F.; Qing, X.; Liu, J.; Zhang, Z.; Shao, Z. LncRNA AFAP1-AS1 Promotes Tumorigenesis and Epithelial-Mesenchymal Transition of Osteosarcoma through RhoC/ROCK1/P38MAPK/Twist1 Signaling Pathway. J. Exp. Clin. Cancer Res. 2019, 38, 375. [Google Scholar] [CrossRef] [Green Version]
  64. Yu, X.J.; Guo, X.Z.; Li, C.; Chong, Y.; Song, T.N.; Pang, J.F.; Shao, M. SIRT1-ZEB1-Positive Feedback Promotes Epithelial-Mesenchymal Transition Process and Metastasis of Osteosarcoma. J. Cell Biochem. 2019, 120, 3727–3735. [Google Scholar] [CrossRef]
  65. Feng, T.; Zhu, Z.; Jin, Y.; Wang, H.; Mao, X.; Liu, D.; Li, Y.; Lu, L.; Zuo, G. The MicroRNA 708 5p/ZEB1/EMT Axis Mediates the Metastatic Potential of Osteosarcoma. Oncol. Rep. 2020, 43, 491–502. [Google Scholar] [CrossRef]
  66. Yao, H.; Hou, G.; Wang, Q.Y.; Xu, W.B.; Zhao, H.Q.; Xu, Y.C. LncRNA SPRY4-IT1 Promotes Progression of Osteosarcoma by Regulating ZEB1 and ZEB2 Expression through Sponging of MiR-101 Activity. Int. J. Oncol. 2020, 56, 85–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Shen, A.; Zhang, Y.; Yang, H.; Xu, R.; Huang, G. Overexpression of ZEB1 Relates to Metastasis and Invasion in Osteosarcoma. J. Surg. Oncol. 2012, 105, 830–834. [Google Scholar] [CrossRef] [PubMed]
  68. Verrecchia, F.; Rédini, F. Transforming Growth Factor-β Signaling Plays a Pivotal Role in the Interplay between Osteosarcoma Cells and Their Microenvironment. Front. Oncol. 2018, 8, 133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Lin, C.Y.; Hsieh, Y.S.; Chu, S.C.; Hsu, L.S.; Huang, S.C.; Chen, P.N. Reduction of Invasion and Cell Stemness and Induction of Apoptotic Cell Death by Cinnamomum Cassia Extracts on Human Osteosarcoma Cells. Environ. Toxicol. 2022, 37, 1261–1274. [Google Scholar] [CrossRef]
  70. He, D.; Gao, J.; Zheng, L.; Liu, S.; Ye, L.; Lai, H.; Pan, B.; Pan, W.; Lou, C.; Chen, Z.; et al. TGF-β Inhibitor RepSox Suppresses Osteosarcoma via the JNK/Smad3 Signaling Pathway. Int. J. Oncol. 2021, 59, 84. [Google Scholar] [CrossRef]
  71. Ma, K.; Zhang, C.; Li, W. Gamabufotalin Suppressed Osteosarcoma Stem Cells through the TGF-β/Periostin/PI3K/AKT Pathway. Chem. Biol. Interact. 2020, 331, 109275. [Google Scholar] [CrossRef]
  72. Jiang, X.; Zhang, Z.; Song, C.; Deng, H.; Yang, R.; Zhou, L.; Sun, Y.; Zhang, Q. Glaucocalyxin A Reverses EMT and TGF-Β1-Induced EMT by Inhibiting TGF-Β1/Smad2/3 Signaling Pathway in Osteosarcoma. Chem. Biol. Interact. 2019, 307, 158–166. [Google Scholar] [CrossRef]
  73. Dong, F.; Liu, T.; Jin, H.; Wang, W. Chimaphilin Inhibits Human Osteosarcoma Cell Invasion and Metastasis through Suppressing the TGF-Β1-Induced Epithelial-to-Mesenchymal Transition Markers via PI-3K/Akt, ERK1/2, and Smad Signaling Pathways. Can. J. Physiol. Pharmacol. 2018, 96, 1–7. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, Y.; Wang, H.; Zhou, R.; Zhong, W.; Lu, S.; Ma, Z.; Chai, Y. Baicalin Inhibits Human Osteosarcoma Cells Invasion, Metastasis, and Anoikis Resistance by Suppressing the Transforming Growth Factor-Β1-Induced Epithelial-to-Mesenchymal Transition. Anticancer Drugs 2017, 28, 581–587. [Google Scholar] [CrossRef] [PubMed]
  75. Kang, H.M.; Park, B.S.; Kang, H.K.; Park, H.R.; Yu, S.B.; Kim, I.R. Delphinidin Induces Apoptosis and Inhibits Epithelial-to-Mesenchymal Transition via the ERK/P38 MAPK-Signaling Pathway in Human Osteosarcoma Cell Lines. Environ. Toxicol. 2018, 33, 640–649. [Google Scholar] [CrossRef] [PubMed]
  76. Xie, C.; Liu, S.; Wu, B.; Zhao, Y.; Chen, B.; Guo, J.; Qiu, S.H.; Cao, Y.M. MiR-19 Promotes Cell Proliferation, Invasion, Migration, and EMT by Inhibiting SPRED2-Mediated Autophagy in Osteosarcoma Cells. Cell Transplant. 2020, 29, 963689720962460. [Google Scholar] [CrossRef]
  77. Raimondi, L.; Gallo, A.; Cuscino, N.; de Luca, A.; Costa, V.; Carina, V.; Bellavia, D.; Bulati, M.; Alessandro, R.; Fini, M.; et al. Potential Anti-Metastatic Role of the Novel MiR-CT3 in Tumor Angiogenesis and Osteosarcoma Invasion. Int. J. Mol. Sci. 2022, 23, 705. [Google Scholar] [CrossRef]
  78. Wang, Z.; Sun, X.; Bao, Y.; Mo, J.; Du, H.; Hu, J.; Zhang, X. E2F1 Silencing Inhibits Migration and Invasion of Osteosarcoma Cells via Regulating DDR1 Expression. Int. J. Oncol. 2017, 51, 1639–1650. [Google Scholar] [CrossRef] [Green Version]
  79. Zheng, B.; Zhou, C.; Qu, G.; Ren, C.; Yan, P.; Guo, W.; Yue, B. VEGFR2 Promotes Metastasis and PD-L2 Expression of Human Osteosarcoma Cells by Activating the STAT3 and RhoA-ROCK-LIMK2 Pathways. Front. Oncol. 2020, 10, 543562. [Google Scholar] [CrossRef]
  80. Han, Y.; Guo, W.; Ren, T.; Huang, Y.; Wang, S.; Liu, K.; Zheng, B.; Yang, K.; Zhang, H.; Liang, X. Tumor-Associated Macrophages Promote Lung Metastasis and Induce Epithelial-Mesenchymal Transition in Osteosarcoma by Activating the COX-2/STAT3 Axis. Cancer Lett. 2019, 440–441, 116–125. [Google Scholar] [CrossRef]
  81. Huang, H.; Han, Y.; Chen, Z.; Pan, X.; Yuan, P.; Zhao, X.; Zhu, H.; Wang, J.; Sun, X.; Shi, P. ML264 Inhibits Osteosarcoma Growth and Metastasis via Inhibition of JAK2/STAT3 and WNT/β-Catenin Signalling Pathways. J. Cell Mol. Med. 2020, 24, 5652–5664. [Google Scholar] [CrossRef] [Green Version]
  82. Hu, Y.; Luo, X.; Zhou, J.; Chen, S.; Gong, M.; Deng, Y.; Zhang, H. Piperlongumine Inhibits the Progression of Osteosarcoma by Downregulating the SOCS3/JAK2/STAT3 Pathway via MiR-30d-5p. Life Sci. 2021, 277, 119501. [Google Scholar] [CrossRef]
  83. Zhang, J.; Li, N.; Lu, S.; Chen, Y.; Shan, L.; Zhao, X.; Xu, Y. The Role of Notch Ligand Jagged1 in Osteosarcoma Proliferation, Metastasis, and Recurrence. J. Orthop. Surg. Res. 2021, 16, 226. [Google Scholar] [CrossRef] [PubMed]
  84. Yu, L.; Xia, K.; Gao, T.; Chen, J.; Zhang, Z.; Sun, X.; Simões, B.M.; Eyre, R.; Fan, Z.; Guo, W.; et al. The Notch Pathway Promotes Osteosarcoma Progression through Activation of Ephrin Reverse Signaling. Mol. Cancer Res. 2019, 17, 2383–2394. [Google Scholar] [CrossRef] [Green Version]
  85. Li, Z.; Tang, Y.; Xing, W.; Dong, W.; Wang, Z. LncRNA, CRNDE Promotes Osteosarcoma Cell Proliferation, Invasion and Migration by Regulating Notch1 Signaling and Epithelial-Mesenchymal Transition. Exp. Mol. Pathol. 2018, 104, 19–25. [Google Scholar] [CrossRef] [PubMed]
  86. Deng, Y.; Zhao, F.; Zhang, Z.; Sun, F.; Wang, M. Long Noncoding RNA SNHG7 Promotes the Tumor Growth and Epithelial-to-Mesenchymal Transition via Regulation of MiR-34a Signals in Osteosarcoma. Cancer Biother Radiopharm. 2018, 33, 365–372. [Google Scholar] [CrossRef] [PubMed]
  87. Dai, G.; Liu, G.; Zheng, D.; Song, Q. Inhibition of the Notch Signaling Pathway Attenuates Progression of Cell Motility, Metastasis, and Epithelial-to-Mesenchymal Transition-like Phenomena Induced by Low Concentrations of Cisplatin in Osteosarcoma. Eur. J. Pharmacol. 2021, 899, 174058. [Google Scholar] [CrossRef] [PubMed]
  88. Ma, Y.; Ren, Y.; Han, E.Q.; Li, H.; Chen, D.; Jacobs, J.J.; Gitelis, S.; O’Keefe, R.J.; Konttinen, Y.T.; Yin, G.; et al. Inhibition of the Wnt-β-Catenin and Notch Signaling Pathways Sensitizes Osteosarcoma Cells to Chemotherapy. Biochem. Biophys. Res. Commun. 2013, 431, 274–279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Wang, S.; Zhang, D.; Han, S.; Gao, P.; Liu, C.; Li, J.; Pan, X. Fibulin-3 Promotes Osteosarcoma Invasion and Metastasis by Inducing Epithelial to Mesenchymal Transition and Activating the Wnt/β-Catenin Signaling Pathway. Sci. Rep. 2017, 7, 6215. [Google Scholar] [CrossRef] [Green Version]
  90. Cao, J.; Han, X.; Qi, X.; Jin, X.; Li, X. TUG1 Promotes Osteosarcoma Tumorigenesis by Upregulating EZH2 Expression via MIR-144-3p. Int. J. Oncol. 2017, 51, 1115–1123. [Google Scholar] [CrossRef] [Green Version]
  91. Zhang, M.; Wang, D.; Zhu, T.; Yin, R. RASSF4 Overexpression Inhibits the Proliferation, Invasion, EMT, and Wnt Signaling Pathway in Osteosarcoma Cells. Oncol. Res. 2017, 25, 83–91. [Google Scholar] [CrossRef] [PubMed]
  92. Yu, M.; Guo, D.; Cao, Z.; Xiao, L.; Wang, G. Inhibitory Effect of MicroRNA-107 on Osteosarcoma Malignancy Through Regulation of Wnt/β-Catenin Signaling in Vitro. Cancer Investig. 2018, 36, 175–184. [Google Scholar] [CrossRef]
  93. Tian, H.; Zhou, T.; Chen, H.; Li, C.; Jiang, Z.; Lao, L.; Kahn, S.A.; Duarte, M.E.L.; Zhao, J.; Daubs, M.D.; et al. Bone Morphogenetic Protein-2 Promotes Osteosarcoma Growth by Promoting Epithelial-Mesenchymal Transition (EMT) through the Wnt/β-Catenin Signaling Pathway. J. Orthop. Res. 2019, 37, 1638–1648. [Google Scholar] [CrossRef] [PubMed]
  94. Fan, S.; Gao, X.; Chen, P.; Li, X. Carboxypeptidase E-ΔN Promotes Migration, Invasiveness, and Epithelial-Mesenchymal Transition of Human Osteosarcoma Cells via the Wnt-β-Catenin Pathway. Biochem. Cell Biol. 2019, 97, 446–453. [Google Scholar] [CrossRef] [PubMed]
  95. Chen, X.; Zhong, L.; Li, X.; Liu, W.; Zhao, Y.; Li, J. Down-Regulation of MicroRNA-31-5p Inhibits Proliferation and Invasion of Osteosarcoma Cells through Wnt/β-Catenin Signaling Pathway by Enhancing AXIN1. Exp. Mol. Pathol. 2019, 108, 32–41. [Google Scholar] [CrossRef]
  96. Liu, Q.; Wang, Z.; Zhou, X.; Tang, M.; Tan, W.; Sun, T.; Deng, Y. MiR-342-5p Inhibits Osteosarcoma Cell Growth, Migration, Invasion, and Sensitivity to Doxorubicin through Targeting Wnt7b. Cell Cycle 2019, 18, 3325–3336. [Google Scholar] [CrossRef] [PubMed]
  97. Zeng, Q.; Li, Z.; Zhao, X.; Guo, L.; Yu, C.; Qin, J.; Zhang, S.; Zhang, Y.; Yang, X. Ubiquitin-Specific Protease 7 Promotes Osteosarcoma Cell Metastasis by Inducing Epithelial-Mesenchymal Transition. Oncol. Rep. 2019, 41, 543–551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Ding, Q.; Mo, F.; Cai, X.; Zhang, W.; Wang, J.; Yang, S.; Liu, X. LncRNA CRNDE Is Activated by SP1 and Promotes Osteosarcoma Proliferation, Invasion, and Epithelial-Mesenchymal Transition via Wnt/β-Catenin Signaling Pathway. J. Cell Biochem. 2020, 121, 3358–3371. [Google Scholar] [CrossRef]
  99. Chen, X.; Zhao, W.; Fan, W. Long Non-Coding RNA GHET1 Promotes Osteosarcoma Development and Progression via Wnt/β-Catenin Signaling Pathway. Oncol. Rep. 2020, 44, 349–359. [Google Scholar] [CrossRef]
  100. Yi, Z.; Pu, Y.; Gou, R.; Chen, Y.; Ren, X.; Liu, W.; Dong, P. Silencing of RIPK4 Inhibits Epithelial-Mesenchymal Transition by Inactivating the Wnt/β-Catenin Signaling Pathway in Osteosarcoma. Mol. Med. Rep. 2020, 21, 1154–1162. [Google Scholar] [CrossRef] [Green Version]
  101. Wang, H.; Zhang, P. LncRNA-CASC15 Promotes Osteosarcoma Proliferation and Metastasis by Regulating Epithelial-Mesenchymal Transition via the Wnt/β-Catenin Signaling Pathway. Oncol. Rep. 2021, 45, 76. [Google Scholar] [CrossRef]
  102. Liang, K.; Liao, L.; Liu, Q.; Ouyang, Q.; Jia, L.; Wu, G. MicroRNA-377-3p Inhibits Osteosarcoma Progression by Targeting CUL1 and Regulating Wnt/β-Catenin Signaling Pathway. Clin. Transl. Oncol. 2021, 23, 2350–2357. [Google Scholar] [CrossRef]
  103. Zhang, H.; Zhou, Q.; Shen, W. Circ-FOXM1 Promotes the Proliferation, Migration and EMT Process of Osteosarcoma Cells through FOXM1-Mediated Wnt Pathway Activation. J. Orthop. Surg. Res. 2022, 17, 344. [Google Scholar] [CrossRef]
  104. Bi, W.; Yang, M.; Xing, P.; Huang, T. MicroRNA MiR-331-3p Suppresses Osteosarcoma Progression via the Bcl-2/Bax and Wnt/β-Catenin Signaling Pathways and the Epithelial-Mesenchymal Transition by Targeting N-Acetylglucosaminyltransferase I (MGAT1). Bioengineered 2022, 13, 14159–14174. [Google Scholar] [CrossRef]
  105. Singla, A.; Wang, J.; Yang, R.; Geller, D.S.; Loeb, D.M.; Hoang, B.H. Wnt Signaling in Osteosarcoma. In Current Advances in Osteosarcoma; Kleinerman, E.S., Ed.; Springer Nature: Cham, Switzerland, 2020; pp. 125–139. ISBN 978-3-319-04843-7. [Google Scholar]
  106. Zhang, D.; Wang, S.; Chen, J.; Liu, H.; Lu, J.; Hua, J.; Huang, A.; Chen, Y. Fibulin-4 Promotes Osteosarcoma Invasion and Metastasis by Inducing Epithelial to Mesenchymal Transition via the PI3K/Akt/MTOR Pathway. Int. J. Oncol. 2017, 50, 1513–1530. [Google Scholar] [CrossRef] [Green Version]
  107. Zhang, X.; Qu, P.; Zhao, H.; Zhao, T.; Cao, N. COX-2 Promotes Epithelial-Mesenchymal Transition and Migration in Osteosarcoma MG-63 Cells via PI3K/AKT/NF-ΚB Signaling. Mol. Med. Rep. 2019, 20, 3811–3819. [Google Scholar] [CrossRef] [Green Version]
  108. Zhang, Y.; Cheng, H.; Li, W.; Wu, H.; Yang, Y. Highly-Expressed P2X7 Receptor Promotes Growth and Metastasis of Human HOS/MNNG Osteosarcoma Cells via PI3K/Akt/GSK3β/β-Catenin and MTOR/HIF1α/VEGF Signaling. Int. J. Cancer 2019, 145, 1068–1082. [Google Scholar] [CrossRef] [Green Version]
  109. Wang, S.; Zhao, G.; Zhao, S.; Qiao, Y.; Yang, H. The Effects of Interleukin-33 (IL-33) on Osteosarcoma Cell Viability, Apoptosis, and Epithelial-Mesenchymal Transition Are Mediated through the PI3K/AKT Pathway. Med. Sci. Monit. 2020, 26, e920766-1–e920766-10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Liu, W.; Jiang, D.; Gong, F.; Huang, Y.; Luo, Y.; Rong, Y.; Wang, J.; Ge, X.; Ji, C.; Fan, J.; et al. MiR-210-5p Promotes Epithelial–Mesenchymal Transition by Inhibiting PIK3R5 Thereby Activating Oncogenic Autophagy in Osteosarcoma Cells. Cell Death Dis. 2020, 11, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Lu, D.-G.; Tang, Q.-L.; Wei, J.-H.; He, F.-Y.; Lu, L.; Tang, Y.-J. Targeting EZH2 by MicroRNA-449a Inhibits Osteosarcoma Cell Proliferation, Invasion and Migration via Regulation of PI3K/AKT Signaling Pathway and Epithelial-Mesenchymal Transition. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 1656–1665. [Google Scholar] [CrossRef]
  112. Xing, S.; Wang, C.; Tang, H.; Guo, J.; Liu, X.; Yi, F.; Liu, G.; Wu, X. Down-Regulation of PDGFRβ Suppresses Invasion and Migration in Osteosarcoma Cells by Influencing Epithelial–Mesenchymal Transition. FEBS Open Bio. 2020, 10, 1748–1757. [Google Scholar] [CrossRef] [PubMed]
  113. Wang, X.; Bian, Z.; Hou, C.; Li, M.; Jiang, W.; Zhu, L. Neuropilin and Tolloid-like 2 Regulates the Progression of Osteosarcoma. Gene 2021, 768, 145292. [Google Scholar] [CrossRef]
  114. Zheng, J.; Liu, C.; Shi, J.; Wen, K.; Wang, X. AIM2 Inhibits the Proliferation, Invasion and Migration, and Promotes the Apoptosis of Osteosarcoma Cells by Inactivating the PI3K/AKT/MTOR Signaling Pathway. Mol. Med. Rep. 2022, 25, 53. [Google Scholar] [CrossRef] [PubMed]
  115. Lu, K.H.; Chen, P.N.; Hsieh, Y.H.; Lin, C.Y.; Cheng, F.Y.; Chiu, P.C.; Chu, S.C.; Hsieh, Y.S. 3-Hydroxyflavone Inhibits Human Osteosarcoma U2OS and 143B Cells Metastasis by Affecting EMT and Repressing u-PA/MMP-2 via FAK-Src to MEK/ERK and RhoA/MLC2 Pathways and Reduces 143B Tumor Growth in Vivo. Food Chem. Toxicol. 2016, 97, 177–186. [Google Scholar] [CrossRef] [PubMed]
  116. Zhang, F.; Yan, T.; Guo, W.; Sun, K.; Wang, S.; Bao, X.; Liu, K.; Zheng, B.; Zhang, H.; Ren, T. Novel Oncogene COPS3 Interacts with Beclin1 and Raf-1 to Regulate Metastasis of Osteosarcoma through Autophagy. J. Exp. Clin. Cancer Res. 2018, 37, 135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Lv, D.-B.; Zhang, J.-Y.; Gao, K.; Yu, Z.-H.; Sheng, W.-C.; Yang, G.; Gao, Y.-Z. MicroRNA-765 Targets MTUS1 to Promote the Progression of Osteosarcoma via Mediating ERK/EMT Pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 4618–4628. [Google Scholar] [CrossRef]
  118. Lin, H.; Hao, Y.; Wan, X.; He, J.; Tong, Y. Baicalein Inhibits Cell Development, Metastasis and EMT and Induces Apoptosis by Regulating ERK Signaling Pathway in Osteosarcoma. J. Recept. Signal. Transduct. 2020, 40, 49–57. [Google Scholar] [CrossRef]
  119. Greenfield, E.M.; Collier, C.D.; Getty, P.J. Receptor Tyrosine Kinases in Osteosarcoma: 2019 Update. In Advances in Experimental Medicine and Biology; Springer: Berlin/Heidelberg, Germany, 2020; Volume 1258, pp. 141–155. [Google Scholar]
  120. U.S. National Library of Medicine List of Clinical Trials Studying Tyrosine Kinase Inhibition in Osteosarcoma. Available online: https://clinicaltrials.gov/ct2/results?cond=osteosarcoma&term=tyrosine+kinase (accessed on 5 February 2023).
  121. Sun, W.; Wang, W.; Lei, J.; Li, H.; Wu, Y. Actin-like Protein 6A Is a Novel Prognostic Indicator Promoting Invasion and Metastasis in Osteosarcoma. Oncol. Rep. 2017, 37, 2405–2417. [Google Scholar] [CrossRef] [Green Version]
  122. Dai, F.; Luo, F.; Zhou, R.; Zhou, Q.; Xu, J.; Zhang, Z.; Xiao, J.; Song, L. Calponin 3 Is Associated with Poor Prognosis and Regulates Proliferation and Metastasis in Osteosarcoma. Aging 2020, 12, 14037–14049. [Google Scholar] [CrossRef]
  123. Wu, Y.; Zhou, W.; Yang, Z.; Li, J.; Jin, Y. MiR-185-5p Represses Cells Growth and Metastasis of Osteosarcoma via Targeting Cathepsin E. Int. J. Toxicol. 2022, 41, 115–125. [Google Scholar] [CrossRef]
  124. Pang, X.; Yin, P.; Han, J.; Wang, Z.; Zheng, F.; Chen, X. CPLA2a Correlates with Metastasis and Poor Prognosis of Osteosarcoma by Facilitating Epithelial-Mesenchymal Transition. Pathol. Res. Pract. 2019, 215, 152398. [Google Scholar] [CrossRef]
  125. Zhao, X.; Li, R.; Wang, Q.; Wu, M.; Wang, Y. Overexpression of Carboxypeptidase X M14 Family Member 2 Predicts an Unfavorable Prognosis and Promotes Proliferation and Migration of Osteosarcoma. Diagn. Pathol. 2019, 14, 118. [Google Scholar] [CrossRef] [Green Version]
  126. Li, X.; Xu, R.; Liu, H.; Fang, K. CUL4A Expression in Pediatric Osteosarcoma Tissues and Its Effect on Cell Growth in Osteosarcoma Cells. Tumor Biol. 2016, 37, 8139–8144. [Google Scholar] [CrossRef]
  127. Ma, Y.; Xu, X.; Luo, M. CXCR6 Promotes Tumor Cell Proliferation and Metastasis in Osteosarcoma through the Akt Pathway. Cell Immunol. 2017, 311, 80–85. [Google Scholar] [CrossRef]
  128. Habel, N.; Stefanovska, B.; Carène, D.; Patiño-Garcia, A.; Lecanda, F.; Fromigué, O. CYR61 Triggers Osteosarcoma Metastatic Spreading via an IGF1Rβ-Dependent EMT-like Process. BMC Cancer 2019, 19, 62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Yuan, X.; Piao, L.; Wang, L.; Han, X.; Tong, L.; Shao, S.; Xu, X.; Zhuang, M.; Liu, Z. Erythrocyte Membrane Protein Band 4.1-like 3 Inhibits Osteosarcoma Cell Invasion through Regulation of Snai1-Induced Epithelial-to-Mesenchymal Transition. Aging 2020, 13, 1947–1961. [Google Scholar] [CrossRef]
  130. Yang, Y.; Chen, J.; Chen, Q. Upregulation of HOXB7 Promotes Proliferation and Metastasis of Osteosarcoma Cells. Mol. Med. Rep. 2017, 16, 2773–2778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Xu, W.; Chen, C.; Xu, R.; Li, Y.; Hu, R.; Li, Z.; Zhu, X. Knockdown of HuR Represses Osteosarcoma Cells Migration, Invasion and Stemness through Inhibition of YAP Activation and Increases Susceptibility to Chemotherapeutic Agents. Biomed. Pharmacother. 2018, 102, 587–593. [Google Scholar] [CrossRef]
  132. Gong, X.; Zheng, X.; Huang, Y.; Song, W.; Chen, G.; Chen, T. Monoacylglycerol Lipase (MAGL) Inhibition Impedes the Osteosarcoma Progression by Regulating Epithelial Mesenchymal Transition. Tohoku J. Exp. Med. 2022, 256, 19–26. [Google Scholar] [CrossRef] [PubMed]
  133. Tang, J.; Shen, L.; Yang, Q.; Zhang, C. Overexpression of Metadherin Mediates Metastasis of Osteosarcoma by Regulating Epithelial-Mesenchymal Transition. Cell Prolif. 2014, 47, 427–434. [Google Scholar] [CrossRef]
  134. Jiang, L.; Jiang, S.; Zhou, W.; Huang, J.; Lin, Y.; Long, H.; Luo, Q. Oxidized Low Density Lipoprotein Receptor 1 Promotes Lung Metastases of Osteosarcomas through Regulating the Epithelial-Mesenchymal Transition. J. Transl. Med. 2019, 17, 369. [Google Scholar] [CrossRef]
  135. Zhai, Q.; Qin, J.; Jin, X.; Sun, X.; Wang, L.; Du, W.; Li, T.; Xiang, X. PADI4 Modulates the Invasion and Migration of Osteosarcoma Cells by Down-Regulation of Epithelial-Mesenchymal Transition. Life Sci. 2020, 256, 117968. [Google Scholar] [CrossRef]
  136. Niinaka, Y.; Harada, K.; Fujimuro, M.; Oda, M.; Haga, A.; Hosoki, M.; Uzawa, N.; Arai, N.; Yamaguchi, S.; Yamashiro, M.; et al. Silencing of Autocrine Motility Factor Induces Mesenchymal-to-Epithelial Transition and Suppression of Osteosarcoma Pulmonary Metastasis. Cancer Res. 2010, 70, 9483–9493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Ren, T.; Zheng, B.; Huang, Y.; Wang, S.; Bao, X.; Liu, K.; Guo, W. Osteosarcoma Cell Intrinsic PD-L2 Signals Promote Invasion and Metastasis via the RhoA-ROCK-LIMK2 and Autophagy Pathways. Cell Death Dis. 2019, 10, 261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Wang, X.; Liang, X.; Liang, H.; Wang, B. SENP1/HIF-1α Feedback Loop Modulates Hypoxia-Induced Cell Proliferation, Invasion, and EMT in Human Osteosarcoma Cells. J. Cell Biochem. 2018, 119, 1819–1826. [Google Scholar] [CrossRef]
  139. Yang, P.; Liu, Y.; Qi, Y.C.; Lian, Z.H. High SENP3 Expression Promotes Cell Migration, Invasion, and Proliferation by Modulating DNA Methylation of E-Cadherin in Osteosarcoma. Technol. Cancer Res. Treat. 2020, 19, 1533033820956988. [Google Scholar] [CrossRef] [PubMed]
  140. Meng, Q.; Ren, C.; Wang, L.; Zhao, Y.; Wang, S. Knockdown of ST6Gal-I Inhibits the Growth and Invasion of Osteosarcoma MG-63 Cells. Biomed. Pharmacother. 2015, 72, 172–178. [Google Scholar] [CrossRef] [PubMed]
  141. Zhou, Y.; Jin, Q.; Xiao, W.; Sun, C. Tankyrase1 Antisense Oligodeoxynucleotides Suppress the Proliferation, Migration and Invasion through Hippo/YAP Pathway in Human Osteosarcoma Cells. Pathol. Res. Pract. 2019, 215, 152381. [Google Scholar] [CrossRef]
  142. Zeng, S.X.; Cai, Q.C.; Guo, C.H.; Zhi, L.Q.; Dai, X.; Zhang, D.F.; Ma, W. High Expression of TRIM29 (ATDC) Contributes to Poor Prognosis and Tumor Metastasis by Inducing Epithelial-Mesenchymal Transition in Osteosarcoma. Oncol. Rep. 2017, 38, 1645–1654. [Google Scholar] [CrossRef] [Green Version]
  143. Chen, Y.; Guo, Y.; Yang, H.; Shi, G.; Xu, G.; Shi, J.; Yin, N.; Chen, D. TRIM66 Overexpresssion Contributes to Osteosarcoma Carcin¬ Ogenesis and Indicates Poor Survival Outcome. Oncotarget 2015, 6, 23708. [Google Scholar] [CrossRef] [Green Version]
  144. Liu, W.; Qiao, R.H.; Wang, D.M.; Huang, X.W.; Li, B.; Wang, D. UHRF1 Promotes Human Osteosarcoma Cell Invasion by Downregulating the Expression of E-Cadherin in an Rb1-Dependent Manner. Mol. Med. Rep. 2016, 13, 315–320. [Google Scholar] [CrossRef] [Green Version]
  145. Song, C.; Liu, W.; Li, J. USP17 Is Upregulated in Osteosarcoma and Promotes Cell Proliferation, Metastasis, and Epithelial-Mesenchymal Transition through Stabilizing SMAD4. Tumor Biol. 2017, 39, 1010428317717138. [Google Scholar] [CrossRef] [Green Version]
  146. Zhang, D.; Jiang, F.; Wang, X.; Li, G. Downregulation of Ubiquitin-Specific Protease 22 Inhibits Proliferation, Invasion, and Epithelial-Mesenchymal Transition in Osteosarcoma Cells. Oncol. Res. 2017, 25, 743–751. [Google Scholar] [CrossRef]
  147. Xu, N.; Wang, L.; Sun, P.; Xu, S.; Fu, S.; Sun, Z. Low Arid1a Expression Correlates with Poor Prognosis and Promotes Cell Proliferation and Metastasis in Osteosarcoma. Pathol. Oncol. Res. 2019, 25, 875–881. [Google Scholar] [CrossRef]
  148. Liu, P.; Yang, P.; Zhang, Z.; Liu, M.; Hu, S. Ezrin/NF-ΚB Pathway Regulates EGF-Induced Epithelial-Mesenchymal Transition (EMT), Metastasis, and Progression of Osteosarcoma. Med. Sci. Monit. 2018, 24, 2098–2108. [Google Scholar] [CrossRef]
  149. Yu, G.-H.; Fu, L.; Chen, J.; Wei, F.; Shi, W.-X. Decreased Expression of Ferritin Light Chain in Osteosarcoma and Its Correlation with Epithelial-Mesenchymal Transition. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 2580–2587. [Google Scholar] [CrossRef]
  150. Zhang, J.; Zhang, Y.; Cheng, S.; Mu, Y.; Liu, Y.; Yi, X.; Jiang, D.; Ding, Y.; Zhuang, R. LAIR-1 Overexpression Inhibits Epithelial-Mesenchymal Transition in Osteosarcoma via GLUT1-Related Energy Metabolism. World J. Surg. Oncol. 2020, 18, 136. [Google Scholar] [CrossRef]
  151. Gao, K.; Yin, J.; Dong, J. Deregulated WWOX Is Involved in a Negative Feedback Loop with MicroRNA-214-3p in Osteosarcoma. Int. J. Mol. Med. 2016, 38, 1850–1856. [Google Scholar] [CrossRef] [Green Version]
  152. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  153. Fedele, V.; Melisi, D. Permissive State of EMT: The Role of Immune Cell Compartment. Front. Oncol. 2020, 10, 587. [Google Scholar] [CrossRef]
  154. Alsaab, H.O.; Sau, S.; Alzhrani, R.; Tatiparti, K.; Bhise, K.; Kashaw, S.K.; Iyer, A.K. PD-1 and PD-L1 Checkpoint Signaling Inhibition for Cancer Immunotherapy: Mechanism, Combinations, and Clinical Outcome. Front. Pharmacol. 2017, 8, 561. [Google Scholar] [CrossRef] [Green Version]
  155. Cano, A.; Pérez-Moreno, M.A.; Rodrigo, I.; Locascio, A.; Blanco, M.J.; del Barrio, M.G.; Portillo, F.; Angela Nieto, M. The Transcription Factor Snail Controls Epithelial-Mesenchymal Transitions by Repressing E-Cadherin Expression. Nat. Cell Biol. 2000, 2, 76–83. [Google Scholar] [CrossRef]
  156. Cech, T.R.; Steitz, J.A. The Noncoding RNA Revolution—Trashing Old Rules to Forge New Ones. Cell 2014, 157, 77–94. [Google Scholar] [CrossRef] [Green Version]
  157. Xu, S.; Gong, Y.; Yin, Y.; Xing, H.; Zhang, N. The Multiple Function of Long Noncoding RNAs in Osteosarcoma Progression, Drug Resistance and Prognosis. Biomed. Pharmacother. 2020, 127, 110141. [Google Scholar] [CrossRef]
  158. Lietz, C.E.; Garbutt, C.; Barry, W.T.; Deshpande, V.; Chen, Y.L.; Lozano-Calderon, S.A.; Wang, Y.; Lawney, B.; Ebb, D.; Cote, G.M.; et al. MicroRNA-MRNA Networks Define Translatable Molecular Outcome Phenotypes in Osteosarcoma. Sci. Rep. 2020, 10, 4409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Xiong, W.; Zhang, Y.; Yu, H. Comprehensive Characterization of Circular RNAs in Osteosarcoma Cell Lines. Cell. Signal. 2020, 71, 109603. [Google Scholar] [CrossRef]
  160. Zhang, Y.; Li, J.; Wang, Y.; Jing, J.; Li, J. The Roles of Circular RNAs in Osteosarcoma. Med. Sci. Monit. 2019, 25, 6378–6382. [Google Scholar] [CrossRef]
  161. Liu, F.; Xing, L.; Zhang, X.; Zhang, X. A Four-Pseudogene Classifier Identified by Machine Learning Serves as a Novel Prognostic Marker for Survival of Osteosarcoma. Genes 2019, 10, 414. [Google Scholar] [CrossRef] [Green Version]
  162. Zhang, Y.; Liu, Z.; Fu, Q.; Wang, X.; Yang, L. Identification of 9-Gene Epithelial-Mesenchymal Transition Related Signature of Osteosarcoma by Integrating Multi Cohorts. Technol. Cancer Res. Treat. 2020, 19, 1533033820980769. [Google Scholar] [CrossRef] [PubMed]
  163. Feng, Z.H.; Zheng, L.; Yao, T.; Tao, S.; Wei, X.A.; Zheng, Z.Y.; Zheng, B.J.; Zhang, X.Y.; Huang, B.; Liu, J.H.; et al. EIF4A3-Induced Circular RNA PRKAR1B Promotes Osteosarcoma Progression by MiR-361-3p-Mediated Induction of FZD4 Expression. Cell Death Dis. 2021, 12, 1025. [Google Scholar] [CrossRef] [PubMed]
  164. Sun, F.; Yu, Z.; Wu, B.; Zhang, H.; Ruan, J. LINC00319 Promotes Osteosarcoma Progression by Regulating the MiR-455-3p/NFIB Axis. J. Gene Med. 2020, 22, e3248. [Google Scholar] [CrossRef] [PubMed]
  165. Wu, S.; Gu, Z.; Wu, Y.; Wu, W.; Mao, B.; Zhao, S. LINC00324 Accelerates the Proliferation and Migration of Osteosarcoma through Regulating WDR66. J. Cell Physiol. 2020, 235, 339–348. [Google Scholar] [CrossRef] [PubMed]
  166. Lian, H.; Xie, P.; Yin, N.; Zhang, J.; Zhang, X.; Li, J.; Zhang, C. Linc00460 Promotes Osteosarcoma Progression via MiR-1224-5p/FADS1 Axis. Life Sci. 2019, 233, 116757. [Google Scholar] [CrossRef]
  167. Bian, X.; Sun, Y.M.; Wang, L.M.; Shang, Y.L. ELK1-Induced Upregulation LncRNA LINC02381 Accelerates the Osteosarcoma Tumorigenesis through Targeting CDCA4 via Sponging MiR-503–5p. Biochem. Biophys. Res. Commun. 2021, 548, 112–119. [Google Scholar] [CrossRef] [PubMed]
  168. Han, G.; Guo, Q.; Ma, N.; Bi, W.; Xu, M.; Jia, J.; Wang, W. LncRNA BCRT1 Facilitates Osteosarcoma Progression via Regulating MiR-1303/FGF7 Axis. Aging 2021, 13, 15501–15510. [Google Scholar] [CrossRef]
  169. Yan, L.; Wu, X.; Yin, X.; Du, F.; Liu, Y.; Ding, X. LncRNA CCAT2 Promoted Osteosarcoma Cell Proliferation and Invasion. J. Cell Mol. Med. 2018, 22, 2592–2599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Zhang, H.; Lin, J.; Chen, J.; Gu, W.; Mao, Y.; Wang, H.; Zhang, Y.; Liu, W. DDX11-AS1 Contributes to Osteosarcoma Progression via Stabilizing DDX11. Life Sci. 2020, 254, 117392. [Google Scholar] [CrossRef] [PubMed]
  171. Wang, Y.; Zhao, Z.; Zhang, S.; Li, Z.; Li, D.; Yang, S.; Zhang, H.; Zeng, X.; Liu, J. LncRNA FAL1 Is a Negative Prognostic Biomarker and Exhibits Pro-Oncogenic Function in Osteosarcoma. J. Cell Biochem. 2018, 119, 8481–8489. [Google Scholar] [CrossRef]
  172. Yang, W.; Shan, Z.; Zhou, X.; Peng, L.; Zhi, C.; Chai, J.; Liu, H.; Yang, J.; Zhang, Z. Knockdown of LncRNA GHET1 Inhibits Osteosarcoma Cells Proliferation, Invasion, Migration and EMT in Vitro and in Vivo. Cancer Biomark. 2018, 23, 589–601. [Google Scholar] [CrossRef]
  173. Zhao, W.; Li, L. SP1-Induced Upregulation of Long Non-Coding RNA HCP5 Promotes the Development of Osteosarcoma. Pathol. Res. Pract. 2019, 215, 439–445. [Google Scholar] [CrossRef]
  174. Cai, L.; Lv, J.; Zhang, Y.; Li, J.; Wang, Y.; Yang, H. The LncRNA HNF1A-AS1 Is a Negative Prognostic Factor and Promotes Tumorigenesis in Osteosarcoma. J. Cell Mol. Med. 2017, 21, 2654–2662. [Google Scholar] [CrossRef] [Green Version]
  175. Lin, H.; Zhao, Z.; Hao, Y.; He, J.; He, J. Long Noncoding RNA HIF1A-AS2 Facilitates Cell Survival and Migration by Sponging MiR-33b-5p to Modulate SIRT6 Expression in Osteosarcoma. Biochem. Cell Biol. 2020, 98, 284–292. [Google Scholar] [CrossRef]
  176. Wang, Y.; Zhang, R.; Cheng, G.; Xu, R.; Han, X. Long Non-Coding RNA HOXA-AS2 Promotes Migration and Invasion by Acting as a CeRNA of MiR-520c-3p in Osteosarcoma Cells. Cell Cycle 2018, 17, 1637–1648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. He, J.W.; Li, D.; Zhou, J.H.; Zhu, Y.L.; Yu, B. qing SP1-Mediated Upregulation of LncRNA LMCD1-AS1 Functions a CeRNA for MiR-106b-5p to Facilitate Osteosarcoma Progression. Biochem. Biophys. Res. Commun. 2020, 526, 670–677. [Google Scholar] [CrossRef] [PubMed]
  178. Li, J.; Wu, Q.M.; Wang, X.Q.; Zhang, C.Q. Long Noncoding RNA MiR210HG Sponges MiR-503 to Facilitate Osteosarcoma Cell Invasion and Metastasis. DNA Cell Biol. 2017, 36, 1117–1125. [Google Scholar] [CrossRef] [PubMed]
  179. Wu, F.; Zhong, Y.; Lang, X.-B.; Tu, Y.-L.; Sun, S.-F. MNX1-AS1 Accelerates the Epithelial-Mesenchymal Transition in Osteosarcoma Cells by Activating MNX1 as a Functional Oncogene. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 8194–8202. [Google Scholar] [CrossRef]
  180. Zhang, L.; Zhao, G.; Ji, S.; Yuan, Q.; Zhou, H. Downregulated Long Non-Coding RNA MSC-AS1 Inhibits Osteosarcoma Progression and Increases Sensitivity to Cisplatin by Binding to MicroRNA-142. Med. Sci. Monit. 2020, 26, e921594-1–e921594-15. [Google Scholar] [CrossRef]
  181. Tan, H.; Zhao, L. LncRNA Nuclear-Enriched Abundant Transcript 1 Promotes Cell Proliferation and Invasion by Targeting MiR-186-5p/HIF-1α in Osteosarcoma. J. Cell Biochem. 2019, 120, 6502–6514. [Google Scholar] [CrossRef]
  182. Liu, W.; Liu, P.; Gao, H.; Wang, X.; Yan, M. Long Non-Coding RNA PGM5-AS1 Promotes Epithelial-Mesenchymal Transition, Invasion and Metastasis of Osteosarcoma Cells by Impairing MiR-140-5p-Mediated FBN1 Inhibition. Mol. Oncol. 2020, 14, 2660–2677. [Google Scholar] [CrossRef]
  183. Xun, C.; Jiang, D.; Tian, Z.; Yunus, A.; Chen, J. Long Noncoding RNA Plasmacytoma Variant Translocation Gene 1 Promotes Epithelial-Mesenchymal Transition in Osteosarcoma. J. Clin. Lab. Anal. 2021, 35, e23587. [Google Scholar] [CrossRef]
  184. Tong, C.-J.; Deng, Q.-C.; Ou, D.-J.; Long, X.; Liu, H.; Huang, K. LncRNA RUSC1-AS1 Promotes Osteosarcoma Progression through Regulating the MiR-340-5p and PI3K/AKT Pathway. Aging 2021, 13, 20116–20130. [Google Scholar] [CrossRef]
  185. Deng, R.; Zhang, J.; Chen, J. LncRNA SNHG1 Negatively Regulates MiRNA-101-3p to Enhance the Expression of ROCK1 and Promote Cell Proliferation, Migration and Invasion in Osteosarcoma. Int. J. Mol. Med. 2019, 43, 1157–1166. [Google Scholar] [CrossRef] [Green Version]
  186. Huang, Y.F.; Lu, L.; Shen, H.L.; Lu, X.X. LncRNA SNHG4 Promotes Osteosarcoma Proliferation and Migration by Sponging MiR-377-3p. Mol. Genet. Genom. Med. 2020, 8, e1349. [Google Scholar] [CrossRef]
  187. Zhang, J.; Ju, C.; Zhang, W.; Xie, L. LncRNA SNHG20 Is Associated with Clinical Progression and Enhances Cell Migration and Invasion in Osteosarcoma. IUBMB Life 2018, 70, 1115–1121. [Google Scholar] [CrossRef] [Green Version]
  188. Yu, X.; Hu, L.; Li, S.; Shen, J.; Wang, D.; Xu, R.; Yang, H. Long Non-Coding RNA Taurine Upregulated Gene 1 Promotes Osteosarcoma Cell Metastasis by Mediating HIF-1α via MiR-143-5p. Cell Death Dis. 2019, 10, 280. [Google Scholar] [CrossRef] [Green Version]
  189. Zhao, X.; Xu, Y.; Sun, X.; Ma, Y.; Zhang, Y.; Wang, Y.; Guan, H.; Jia, Z.; Li, Y.; Wang, Y. MiR-17-5p Promotes Proliferation and Epithelial-Mesenchymal Transition in Human Osteosarcoma Cells by Targeting SRC Kinase Signaling Inhibitor 1. J. Cell Biochem. 2019, 120, 5495–5504. [Google Scholar] [CrossRef]
  190. Zhang, H.; Zhang, J.; Meng, F.; Zhu, H.; Yan, H.; Guo, Y.; Zhang, S. MicroRNA-93 Promotes the Tumorigenesis of Osteosarcoma by Targeting TIMP2. Biosci. Rep. 2019, 39, BSR20191237. [Google Scholar] [CrossRef] [Green Version]
  191. Chen, J.; Yan, D.; Wu, W.; Zhu, J.; Ye, W.; Shu, Q. Micro RNA-130a Promotes the Metastasis and Epithelialmesenchymal Transition of Osteosarcoma by Targeting PTEN. Oncol. Rep. 2016, 35, 3285–3292. [Google Scholar] [CrossRef] [Green Version]
  192. Shen, S.; Huang, K.; Wu, Y.; Ma, Y.; Wang, J.; Qin, F.; Ma, J. A MiR-135b-TAZ Positive Feedback Loop Promotes Epithelial–Mesenchymal Transition (EMT) and Tumorigenesis in Osteosarcoma. Cancer Lett. 2017, 407, 32–44. [Google Scholar] [CrossRef]
  193. Yao, J.; Lin, J.; He, L.; Huang, J.; Liu, Q. TNF-α/MiR-155 Axis Induces the Transformation of Osteosarcoma Cancer Stem Cells Independent of TP53INP1. Gene 2020, 726, 144224. [Google Scholar] [CrossRef]
  194. Wang, X.; Zhang, L.; Zhang, X.; Xing, C.; Liu, R.; Zhang, F. MiR-196a Promoted Cell Migration, Invasion and the Epithelial-Mesenchymal Transition by Targeting HOXA5 in Osteosarcoma. Cancer Biomark. 2020, 29, 291–298. [Google Scholar] [CrossRef]
  195. Chen, Z.; Zhao, G.; Zhang, Y.; Ma, Y.; Ding, Y.; Xu, N. MiR-199b-5p Promotes Malignant Progression of Osteosarcoma by Regulating HER2. J. BUON 2018, 23, 1816–1824. [Google Scholar]
  196. Shi, C.; Huang, C.M.; Wang, B.; Sun, T.F.; Zhu, A.X.; Zhu, Y.C. Pseudogene MSTO2P Enhances Hypoxia-Induced Osteosarcoma Malignancy by Upregulating PD-L1. Biochem. Biophys. Res. Commun. 2020, 530, 673–679. [Google Scholar] [CrossRef]
  197. Ma, L.; Zhang, L.; Guo, A.; Liu, L.C.; Yu, F.; Diao, N.; Xu, C.; Wang, D. Overexpression of FER1L4 Promotes the Apoptosis and Suppresses Epithelial-Mesenchymal Transition and Stemness Markers via Activating PI3K/AKT Signaling Pathway in Osteosarcoma Cells. Pathol. Res. Pract. 2019, 215, 152412. [Google Scholar] [CrossRef]
  198. Ye, F.; Tian, L.; Zhou, Q.; Feng, D. LncRNA FER1L4 Induces Apoptosis and Suppresses EMT and the Activation of PI3K/AKT Pathway in Osteosarcoma Cells via Inhibiting MiR-18a-5p to Promote SOCS5. Gene 2019, 721, 144093. [Google Scholar] [CrossRef]
  199. Ye, K.; Wang, S.; Zhang, H.; Han, H.; Ma, B.; Nan, W. Long Noncoding RNA GAS5 Suppresses Cell Growth and Epithelial–Mesenchymal Transition in Osteosarcoma by Regulating the MiR-221/ARHI Pathway. J. Cell Biochem. 2017, 118, 4772–4781. [Google Scholar] [CrossRef]
  200. Shen, B.; Zhou, N.; Hu, T.; Zhao, W.; Wu, D.; Wang, S. LncRNA MEG3 Negatively Modified Osteosarcoma Development through Regulation of MiR-361-5p and FoxM1. J. Cell Physiol. 2019, 234, 13464–13480. [Google Scholar] [CrossRef]
  201. Zhang, G.; Li, Y.; Liao, G.; Qiu, H. LncRNA NKILA Inhibits Invasion and Migration of Osteosarcoma Cells via NF-ΚB/Snail Signaling Pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 4118–4125. [Google Scholar]
  202. Fan, H.; Liu, T.; Tian, H.; Zhang, S. Tusc8 Inhibits the Development of Osteosarcoma by Sponging Mir-197-3p and Targeting Ehd2. Int. J. Mol. Med. 2020, 46, 1311–1320. [Google Scholar] [CrossRef]
  203. Zhang, Z.; Zhao, M.; Wang, G. Upregulation of MicroRNA-7 Contributes to Inhibition of the Growth and Metastasis of Osteosarcoma Cells through the Inhibition of IGF1R. J. Cell Physiol. 2019, 234, 22195–22206. [Google Scholar] [CrossRef]
  204. Jiao, Z.-H.; Wang, J.-D.; Wang, X.-J. MicroRNA-16 Suppressed the Invasion and Migration of Osteosarcoma by Directly Inhibiting RAB23. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 2598–2605. [Google Scholar] [CrossRef]
  205. Chen, B.; Liu, J.; Qu, J.; Song, Y.; Li, Y.; Pan, S. MicroRNA-25 Suppresses Proliferation, Migration, and Invasion of Osteosarcoma by Targeting SOX4. Tumor Biol. 2017, 39, 1010428317703841. [Google Scholar] [CrossRef] [Green Version]
  206. Gong, H.L.; Tao, Y.; Mao, X.Z.; Song, D.Y.; You, D.; Ni, J.D. MicroRNA-29a Suppresses the Invasion and Migration of Osteosarcoma Cells by Regulating the SOCS1/NF-ΚB Signalling Pathway through Negatively Targeting DNMT3B. Int. J. Mol. Med. 2019, 44, 1219–1232. [Google Scholar] [CrossRef] [Green Version]
  207. Waresijiang, N.; Sun, J.; Abuduaini, R.; Jiang, T.; Zhou, W.; Yuan, H. The Downregulation of MIR-125a-5p Functions as a Tumor Suppressor by Directly Targeting MMP-11 in Osteosarcoma. Mol. Med. Rep. 2016, 13, 4859–4864. [Google Scholar] [CrossRef] [Green Version]
  208. Liu, X.; Liang, Z.; Gao, K.; Li, H.; Zhao, G.; Wang, S.; Fang, J. MicroRNA-128 Inhibits EMT of Human Osteosarcoma Cells by Directly Targeting Integrin A2. Tumor Biol. 2016, 37, 7951–7957. [Google Scholar] [CrossRef]
  209. Liu, Y.; Li, Y.; Liu, J.; Wu, Y.; Zhu, Q. MicroRNA-132 Inhibits Cell Growth and Metastasis in Osteosarcoma Cell Lines Possibly by Targeting Sox4. Int. J. Oncol. 2015, 47, 1672–1684. [Google Scholar] [CrossRef] [Green Version]
  210. Shi, Y.K.; Guo, Y.H. MiR-139-5p Suppresses Osteosarcoma Cell Growth and Invasion through Regulating DNMT1. Biochem. Biophys. Res. Commun. 2018, 503, 459–466. [Google Scholar] [CrossRef]
  211. Guo, Q.; Zhang, N.; Liu, S.; Pang, Z.; Chen, Z. By Targeting TRAF6, MiR-140-3p Inhibits TGF-Β1-Induced Human Osteosarcoma Epithelial-to-Mesenchymal Transition, Migration, and Invasion. Biotechnol. Lett. 2020, 42, 2123–2133. [Google Scholar] [CrossRef]
  212. Fu, Y.; Tang, Y.; Wang, J.; Guo, Z. MicroRNA-181c Suppresses the Biological Progression of Osteosarcoma via Targeting Smad7 and Regulating Transforming Growth Factor-β (TGF-β) Signaling Pathway. Med. Sci. Monit. 2019, 25, 4801–4810. [Google Scholar] [CrossRef]
  213. Yang, D.; Liu, G.; Wang, K. MiR-203 Acts as a Tumor Suppressor Gene in Osteosarcoma by Regulating RAB22A. PLoS ONE 2015, 10, e0132225. [Google Scholar] [CrossRef]
  214. He, F.; Fang, L.; Yin, Q. MiR-363 Acts as a Tumor Suppressor in Osteosarcoma Cells by Inhibiting PDZD2. Oncol. Rep. 2019, 41, 2729–2738. [Google Scholar] [CrossRef]
  215. Zhang, Y.; Wang, F.; Wang, L.; Zhang, Q. MiR-363 Suppresses Cell Migration, Invasion, and Epithelial-Mesenchymal Transition of Osteosarcoma by Binding to NOB1. World J. Surg. Oncol. 2020, 18, 83. [Google Scholar] [CrossRef]
  216. Xu, M.; Jin, H.; Xu, C.X.; Sun, B.; Song, Z.G.; Bi, W.Z.; Wang, Y. MiR-382 Inhibits Osteosarcoma Metastasis and Relapse by Targeting y Box-Binding Protein 1. Mol. Ther. 2015, 23, 89–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Tan, Y.; Chen, L.; Li, S.; Hao, H.; Zhang, D. MiR-384 Inhibits Malignant Biological Behavior Such as Proliferation and Invasion of Osteosarcoma by Regulating IGFBP3. Technol. Cancer Res. Treat. 2020, 19, 1533033820909125. [Google Scholar] [CrossRef] [PubMed]
  218. Liu, Y.; Zhang, J.; Xing, C.; Wei, S.; Guo, N.; Wang, Y. MiR-486 Inhibited Osteosarcoma Cells Invasion and Epithelial-Mesenchymal Transition by Targeting PIM1. Cancer Biomark. 2018, 23, 269–277. [Google Scholar] [CrossRef] [PubMed]
  219. Qiu, J.; Zhang, Y.; Chen, H.; Guo, Z. MicroRNA-488 Inhibits Proliferation, Invasion and EMT in Osteosarcoma Cell Lines by Targeting Aquaporin 3. Int. J. Oncol. 2018, 53, 1493–1504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Wang, Y.; Lin, S.; Chen, L.; Qiu, H.; Wang, J. MicroRNA-489 Suppresses Osteosarcoma Invasion, Migration and Epithelial-to-Mesenchymal Transition by Directly Targeting NAA10. Minerva Endocrinol. 2020, 45, 150–153. [Google Scholar] [CrossRef]
  221. Wang, T.; Wang, D.; Zhang, L.; Yang, P.; Wang, J.; Liu, Q.; Yan, F.; Lin, F. The TGFβ-MiR-499a-SHKBP1 Pathway Induces Resistance to EGFR Inhibitors in Osteosarcoma Cancer Stem Cell-like Cells. J. Exp. Clin. Cancer Res. 2019, 38, 226. [Google Scholar] [CrossRef] [Green Version]
  222. Guo, X.; Zhang, J.; Pang, J.; He, S.; Li, G.; Chong, Y.; Li, C.; Jiao, Z.; Zhang, S.; Shao, M. MicroRNA-503 Represses Epithelial–Mesenchymal Transition and Inhibits Metastasis of Osteosarcoma by Targeting c-Myb. Tumor Biol. 2016, 37, 9181–9187. [Google Scholar] [CrossRef]
  223. Wang, D.; Bao, F.; Teng, Y.; Li, Q.; Li, J. MicroRNA-506-3p Initiates Mesenchymal-to-Epithelial Transition and Suppresses Autophagy in Osteosarcoma Cells by Directly Targeting SPHK1. Biosci. Biotechnol. Biochem. 2019, 83, 836–844. [Google Scholar] [CrossRef]
  224. Wang, X.; Li, C.; Yao, W.; Tian, Z.; Liu, Z.; Ge, H. MicroRNA-761 Suppresses Tumor Progression in Osteosarcoma via Negatively Regulating ALDH1B1. Life Sci. 2020, 262, 118544. [Google Scholar] [CrossRef]
  225. di Fiore, R.; Drago-Ferrante, R.; Pentimalli, F.; di Marzo, D.; Forte, I.M.; Carlisi, D.; de Blasio, A.; Tesoriere, G.; Giordano, A.; Vento, R. Let-7d MiRNA Shows Both Antioncogenic and Oncogenic Functions in Osteosarcoma-Derived 3AB-OS Cancer Stem Cells. J. Cell Physiol. 2016, 231, 1832–1841. [Google Scholar] [CrossRef]
  226. Yang, H.; Zhao, L.; Zhang, Y.; Li, F.F. A Comprehensive Analysis of Immune Infiltration in the Tumor Microenvironment of Osteosarcoma. Cancer Med. 2021, 10, 5696–5711. [Google Scholar] [CrossRef]
  227. Ling, J.; Sun, Y.; Pan, J.; Wang, H.; Ma, Z.; Yin, J.; Bao, Z.; Yang, H.; Liu, L. Feedback Modulation of Endothelial Cells Promotes Epithelial-Mesenchymal Transition and Metastasis of Osteosarcoma Cells by Von Willebrand Factor Release. J. Cell Biochem. 2019, 120, 15971–15979. [Google Scholar] [CrossRef]
  228. Dai, J.; Qin, L.; Chen, Y.; Wang, H.; Lin, G.; Li, X.; Liao, H.; Fang, H. Matrix Stiffness Regulates Epithelial-Mesenchymal Transition via Cytoskeletal Remodeling and MRTF-A Translocation in Osteosarcoma Cells. J. Mech. Behav. Biomed. Mater. 2019, 90, 226–238. [Google Scholar] [CrossRef]
  229. Bielack, S.; Kempf-Bielack, B.; Delling, G.; Exner, G.; Flege, S.; Helmke, K.; Kotz, R.; Salzer-Kuntschik, M.; Werner, M.; Winkelmann, W.; et al. Prognostic Factors in High-Grade Osteosarcoma of the Extremities of Trunk: An Analysis of 1,702 Patients Treated on Neoadjuvant Coperative Osteosarcoma Study Group Protocols. J. Clin. Oncol. 2002, 20, 776–790. [Google Scholar] [CrossRef]
  230. de Las Rivas, J.; Brozovic, A.; Izraely, S.; Casas-Pais, A.; Witz, I.P.; Figueroa, A. Cancer Drug Resistance Induced by EMT: Novel Therapeutic Strategies. Arch. Toxicol. 2021, 95, 2279–2297. [Google Scholar] [CrossRef]
  231. Ding, L.; Wang, C.; Cui, Y.; Han, X.; Zhou, Y.; Bai, J.; Li, R. S-Phase Kinase-Associated Protein 2 Is Involved in Epithelial-Mesenchymal Transition in Methotrexate-Resistant Osteosarcoma Cells. Int. J. Oncol. 2018, 52, 1841–1852. [Google Scholar] [CrossRef] [Green Version]
  232. Huang, Y.; Zhang, J.; Shao, H.; Liu, J.; Jin, M.; Chen, J.; Zhao, H. MiR-33a Mediates the Anti-Tumor Effect of Lovastatin in Osteosarcoma by Targeting CYR61. Cell. Physiol. Biochem. 2018, 51, 938–948. [Google Scholar] [CrossRef]
  233. Cheng, H.-L.; Lin, C.-W.; Yang, J.-S.; Hsieh, M.-J.; Yang, S.-F.; Lu, K.-H. Zoledronate Blocks Geranylgeranylation Not Farnesylation to Suppress Human Osteosarcoma U2OS Cells Metastasis by EMT via Rho A Activation and FAK-Inhibited JNK and P38 Pathways. Oncotarget 2016, 7, 9742–9758. [Google Scholar] [CrossRef] [Green Version]
  234. Kim, E.H.; Kim, M.-S.; Lee, K.-H.; Koh, J.-S.; Jung, W.-G.; Kong, C.-B. Zoledronic Acid Is an Effective Radiosensitizer in the Treatment of Osteosarcoma. Oncotarget 2016, 7, 70869–70880. [Google Scholar] [CrossRef] [Green Version]
  235. Fang, D.; Yang, H.; Lin, J.; Teng, Y.; Jiang, Y.; Chen, J.; Li, Y. 17β-Estradiol Regulates Cell Proliferation, Colony Formation, Migration, Invasion and Promotes Apoptosis by Upregulating MiR-9 and Thus Degrades MALAT-1 in Osteosarcoma Cell MG-63 in an Estrogen Receptor-Independent Manner. Biochem. Biophys. Res. Commun. 2015, 457, 500–506. [Google Scholar] [CrossRef]
  236. Sánchez-Sánchez, A.M.; Turos-Cabal, M.; Puente-Moncada, N.; Herrera, F.; Rodríguez, C.; Martín, V. Calcium Acts as a Central Player in Melatonin Antitumor Activity in Sarcoma Cells. Cell. Oncol. 2022, 45, 415–428. [Google Scholar] [CrossRef] [PubMed]
  237. Qu, H.; Xue, Y.; Lian, W.; Wang, C.; He, J.; Fu, Q.; Zhong, L.; Lin, N.; Lai, L.; Ye, Z.; et al. Melatonin Inhibits Osteosarcoma Stem Cells by Suppressing SOX9-Mediated Signaling. Life Sci. 2018, 207, 253–264. [Google Scholar] [CrossRef] [PubMed]
  238. Fontanella, R.; Pelagalli, A.; Nardelli, A.; D’Alterio, C.; Ieranò, C.; Cerchia, L.; Lucarelli, E.; Scala, S.; Zannetti, A. A Novel Antagonist of CXCR4 Prevents Bone Marrow-Derived Mesenchymal Stem Cell-Mediated Osteosarcoma and Hepatocellular Carcinoma Cell Migration and Invasion. Cancer Lett. 2016, 370, 100–107. [Google Scholar] [CrossRef]
  239. Zheng, B.; Ren, T.; Huang, Y.; Guo, W. Apatinib Inhibits Migration and Invasion as Well as PD-L1 Expression in Osteosarcoma by Targeting STAT3. Biochem. Biophys. Res. Commun. 2018, 495, 1695–1701. [Google Scholar] [CrossRef]
  240. Seba, V.; Silva, G.; dos Santos, M.B.; Baek, S.J.; França, S.d.C.; Fachin, A.L.; Regasini, L.O.; Marins, M. Chalcone Derivatives 4′-Amino-1-Naphthyl-Chalcone (D14) and 4′-Amino-4-Methyl-1-Naphthyl-Chalcone (D15) Suppress Migration and Invasion of Osteosarcoma Cells Mediated by P53 Regulating Emt-Related Genes. Int. J. Mol. Sci. 2018, 19, 2838. [Google Scholar] [CrossRef] [Green Version]
  241. Mishra, R.; Nathani, S.; Varshney, R.; Sircar, D.; Roy, P. Berberine Reverses Epithelial-Mesenchymal Transition and Modulates Histone Methylation in Osteosarcoma Cells. Mol. Biol. Rep. 2020, 47, 8499–8511. [Google Scholar] [CrossRef]
  242. Wang, D.; Zhang, K.; Du, G.; Wang, J.; Zhao, J. Berberine Enhances the Radiosensitivity of Osteosarcoma by Targeting Rad51 and Epithelial-Mesenchymal Transition. J. Cancer Res. Ther. 2020, 16, 215–221. [Google Scholar] [CrossRef]
  243. Liu, X.; Fan, Y.; Xie, J.; Zhang, L.; Li, L.; Wang, Z. Dehydroandrographolide Inhibits Osteosarcoma Cell Growth and Metastasis by Targeting SATB2-Mediated EMT. Anticancer Agents Med. Chem. 2019, 19, 1728–1736. [Google Scholar] [CrossRef]
  244. Wang, Y.; Shang, G.; Wang, W.; Qiu, E.; Pei, Y.; Zhang, X. Magnoflorine Inhibits the Malignant Phenotypes and Increases Cisplatin Sensitivity of Osteosarcoma Cells via Regulating MiR-410-3p/HMGB1/NF-ΚB Pathway. Life Sci. 2020, 256, 117967. [Google Scholar] [CrossRef]
  245. Chang, J.; Wang, H.; Wang, X.; Zhao, Y.; Zhao, D.; Wang, C.; Li, Y.; Yang, Z.; Lu, S.; Zeng, Q.; et al. Molecular Mechanisms of Polyphyllin I-Induced Apoptosis and Reversal of the Epithelial-Mesenchymal Transition in Human Osteosarcoma Cells. J. Ethnopharmacol. 2015, 170, 117–127. [Google Scholar] [CrossRef]
  246. Ma, Z.; Yang, J.; Yang, Y.; Wang, X.; Chen, G.; Shi, A.; Lu, Y.; Jia, S.; Kang, X.; Lu, L. Rosmarinic Acid Exerts an Anticancer Effect on Osteosarcoma Cells by Inhibiting DJ-1 via Regulation of the PTEN-PI3K-Akt Signaling Pathway. Phytomedicine 2020, 68, 153186. [Google Scholar] [CrossRef] [PubMed]
  247. Muscella, A.; Stefàno, E.; de Bellis, L.; Nutricati, E.; Negro, C.; Marsigliante, S. Antitumor and Antimigration Effects of Salvia Clandestina L. Extract on Osteosarcoma Cells. Ann. N. Y. Acad. Sci. 2021, 1500, 34–47. [Google Scholar] [CrossRef] [PubMed]
  248. Zhou, D.; He, L. Sauchinone Inhibits Hypoxia-Induced Invasion and Epithelial–Mesenchymal Transition in Osteosarcoma Cells via Inactivation of the Sonic Hedgehog Pathway. J. Recept. Signal. Transduct. 2022, 42, 173–179. [Google Scholar] [CrossRef] [PubMed]
  249. Jiang, C.; Fang, X.; Zhang, H.; Wang, X.; Li, M.; Jiang, W.; Tian, F.; Zhu, L.; Bian, Z. Triptolide Inhibits the Growth of Osteosarcoma by Regulating MicroRNA-181a via Targeting PTEN Gene in Vivo and Vitro. Tumor Biol. 2017, 39, 1010428317697556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  250. Oh, J.Y.; Lee, Y.J.; Kim, E.H. Tumor-Treating Fields Inhibit the Metastatic Potential of Osteosarcoma Cells. Technol. Cancer Res. Treat. 2020, 19, 1533033820947481. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The epithelial to mesenchymal transition (EMT) and the reverse process of the mesenchymal to epithelial transition (MET). EMT is characterized by a loss of epithelial cell markers, an increase in mesenchymal cell markers, a loss of apical cell polarity, a loss of tight cell junctions, and an increased capacity for cell migration and invasion. MET is characterized by a loss of mesenchymal cell markers, an increase in epithelial cell markers, increased apical cell polarity, tight junctions, adherence to a basement membrane, and increased cell proliferation.
Figure 1. The epithelial to mesenchymal transition (EMT) and the reverse process of the mesenchymal to epithelial transition (MET). EMT is characterized by a loss of epithelial cell markers, an increase in mesenchymal cell markers, a loss of apical cell polarity, a loss of tight cell junctions, and an increased capacity for cell migration and invasion. MET is characterized by a loss of mesenchymal cell markers, an increase in epithelial cell markers, increased apical cell polarity, tight junctions, adherence to a basement membrane, and increased cell proliferation.
Biomolecules 13 00398 g001
Figure 2. Signaling pathways in EMT. EMT regulation is complex and affected by multiple pathways, which also interact with each other. Regulation is typically via the Transforming Growth Factor β (TGFβ)/SMAD, Notch, canonical wnt, phosphoinositide 3-kinase (PI3K)/Akt, RAS/RAF, and JAK/STAT pathways. The transcription factors that mediate EMT are primarily Snail, Slug, ZEB1, and TWIST. EMT is characterized by an increased production of N-cadherin, vimentin, and fibronectin, and MET is characterized by an increased production of E-cadherin and Occludin.
Figure 2. Signaling pathways in EMT. EMT regulation is complex and affected by multiple pathways, which also interact with each other. Regulation is typically via the Transforming Growth Factor β (TGFβ)/SMAD, Notch, canonical wnt, phosphoinositide 3-kinase (PI3K)/Akt, RAS/RAF, and JAK/STAT pathways. The transcription factors that mediate EMT are primarily Snail, Slug, ZEB1, and TWIST. EMT is characterized by an increased production of N-cadherin, vimentin, and fibronectin, and MET is characterized by an increased production of E-cadherin and Occludin.
Biomolecules 13 00398 g002
Table 3. Effects of highly expressed non-coding ribonucleic acids.
Table 3. Effects of highly expressed non-coding ribonucleic acids.
Ribonucleic AcidIncreased Levels in Clinical SamplePromoted EMTPromoted Cell Migration/InvasionPromoted In Vivo Tumor GrowthPromoted In Vivo MetastasisAssociated Pathways/Targets
circ-FOXM1 [103]No miR-320a, wnt
circ-PRKAR1B [163]No miR-361-3p, FZD4
LINC00319 [164]Yes miR-455-3p, NFIB
LINC00324 [165]Yes WDR66, HuR
LINC00460 [166]No miR-1224-5p, FADS1
LINC02381 [167]Yes miR-503-5p, CDCA4
lncRNA AFAP1-AS1 [63]Yes Rho, ROCK, p38
lncRNA BCRT1 [168]Yes miR-1303, FGF7
lncRNA CASC15 [101]Yes wnt/β-catenin
lncRNA CCAT2 [169]Yes LATS2, c-Myc
lncRNA CRNDE [85,98]Yes Notch1, SP1, wnt/β-catenin
lncRNA DDX11-AS1 [170]No miR-873-5p, IGF2BP2
lncRNA FAL1 [171]Yes GSK-3β
lncRNA GHET1 [99,172]Yes Ki67, wnt/β-catenin
lncRNA HCP5 [173]No SP1
lncRNA HNF1A-AS1 [174]Yes
lncRNA HIF1A-AS2 [175]Yes miR-33b-5p, SIRT6
lncRNA HOXA-AS2 [176]Yes miR-502c-3p
lncRNA LMCD1-AS1 [177]Yes miR-106b-5p, SP1
lncRNA miR210HG [178]Yes miR-503
lncRNA MNX1-AS1 [179]Yes Snail
lncRNA MSC-AS1 [180]Yes miR-142, CDK6, PI3K/Akt
lncRNA NEAT1 [181]Yes miR-186-5p, HIF-1α
lncRNA PGM5-AS1 [182]Yes miR-140-5p, FBN1
lncRNA PVT1 [183]Yes
lncRNA RUSC1-AS1 [184]Yes miR-340-5p, PI3K/Akt
lncRNA SNHG1 [185]Yes miRNA-101-3p, ROCK1, PI3K/Akt
lncRNA SNHG4 [186]Yes miR-377-3p
lncRNA SNHG7 [86]Yes MiR-34a, Notch-1, BCL-2, CDK6, SMAD4
lncRNA SNHG20 [187]Yes
lncRNA SPRY4-IT1 [47,66]No miR-101
lncRNA TUG1 [90,188]Yes miR-144-3p, miR-143-5p, EZH2, HIF-1α, wnt
lncRNA XIST [57]Yes miR-153, SNAI1
miR-17-5p [189]Yes SRCIN1
miR-19 [76]Yes SPRED2, ERK/MAPK
miR-31-5p [95]Yes AXIN1, wnt/β-catenin
miR-93 [190]Yes TIMP2
miR-130a [191]Yes PTEN
miR-135b [192]No TAZ
miR-155 [193]No TNFa, TP53INP1
miR-196a [194]Yes HOXA5
miR-199b-5p [195]Yes HER2
miR-210-5p [110]Yes PIK3R5, Akt
Pseudogene MSTO2P [196]Yes PD-L1
Biomolecules 13 00398 i005: Significant association; Biomolecules 13 00398 i006: Not studied/reported.
Table 4. Effects of poorly expressed non-coding ribonucleic acids.
Table 4. Effects of poorly expressed non-coding ribonucleic acids.
Ribonucleic AcidDecreased Levels in Clinical SamplesInhibited EMTInhibited Cell Migration/InvasionInhibited In Vivo Tumor GrowthInhibited In Vivo MetastasisAssociated Pathways/Targets
lncRNA FER1L4 [197,198]Yes miR-18a-5p, PI3K/Akt
lncRNA GAS5 [199]Yes miR-221, ARHI
lncRNA MEG3 [200]Yes miR-361-5p, FoxM1
lncRNA NKILA [201]Yes NFκB, Snail
lncRNA TUSC8 [202]Yes miR-197-3p, EHD2
miR-7 [203]Yes IGF1R
miR-16 [204]Yes RAB23
miR-25 [205]Yes SOX4
miR-29a [206]Yes SOCS1/NFκB, DNMT3B
miR-107 [92]Yes wnt/β-catenin
miR-125a-5p [207]Yes MMP11
miR-128 [208]Yes Integrin A2
miR-132 [209]No SOX4
miR-139-5p [210]Yes DNMT1
miR-140-3p [211]Yes TRAF6, TGFB
miR-145 [51]Yes Snail
miR-181c [212]Yes SMAD7, TGFB
miR-203 [213]Yes RAB22A
miR-331-3p [104]Yes MGAT1, Bcl/Bax, wnt/β-catenin
miR-342-5p [96]Yes wnt/β-catenin
miR-363 [214,215]Yes PDZD2, NOB1
miR-377-3p [186]Yes CuL1, wnt/β-catenin
miR-382 [216]Yes YB-1
miR-384 [217]Yes MECP2, IGFBP3
miR-449a [111]Yes EZH2, PI3K/Akt
miR-486 [218]Yes PIM1
miR-488 [219]Yes AQP3
miR-489 [220]Yes NAA10
miR-499a [221]Yes TGFβ, EGFR, Akt, SHKBP1
miR-503 [222]No c-myc
miR-506-3p [223]No SPHK1, LC3II/I
miR-708-5p [65]No ZEB1
miR-761 [224]Yes ALDH1B1, TGFB
miR-765 [117]Yes MTUS, ERK
miR-CT3 [77]Yes p38/MAPK
miR-let-7d [225]No CCND2, E2F2
Biomolecules 13 00398 i007: Significant association; ↑: Inverse association; Biomolecules 13 00398 i008:Not studied/reported.
Table 5. Traditional and herbal medicine effects on EMT in OS.
Table 5. Traditional and herbal medicine effects on EMT in OS.
CompoundInhibits EMTInhibits Cell Migration/InvasionInhibits In Vivo Tumor GrowthInhibits In Vivo MetastasisAssociated Pathways/Targets
3’hydroxyflavone [115] MEK/ERK
Baicalin [74,118] ERK, TGF-β
Berberine [241,242] EZH2, Rad51
Chimaphilin [73] PI3K/Akt, ERK, TGF-β
Cinnamomum cassia extract [69] TGF-β
Dehydroandrogranpholide [243] SATB2
Delphinidin [75] ERK, MAPK
Gamabufotalin [71] PI3K/Akt, TGF-β
Glaucocalyxin A [72] TGF-β, Smad
Magnoflorine [244] miR-410-3p, HMGB1, NF-κB
Nitidine Chloride [46] Akt, GSK-3β, Snail
Oridonin [58] TGF-β, Smad, Snail
Piperlongumine [82] miR-30d-5p, SOCS3, JAK2/STAT3
Polyphillin I [245] NF-κB, c-Myc
Rosmarinic acid [246] DJ-1, PI3K/Akt
Salvia 13landestine extract [247] Akt/PKB
Sauchinone [248] Sonic hedgehog
Triptolide [249] miR-181a, PTEN
Biomolecules 13 00398 i009: Significant association; ↑: Inverse association; Biomolecules 13 00398 i010: Not studied/reported.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hinton, K.; Kirk, A.; Paul, P.; Persad, S. Regulation of the Epithelial to Mesenchymal Transition in Osteosarcoma. Biomolecules 2023, 13, 398. https://doi.org/10.3390/biom13020398

AMA Style

Hinton K, Kirk A, Paul P, Persad S. Regulation of the Epithelial to Mesenchymal Transition in Osteosarcoma. Biomolecules. 2023; 13(2):398. https://doi.org/10.3390/biom13020398

Chicago/Turabian Style

Hinton, Kristin, Andrew Kirk, Paulose Paul, and Sujata Persad. 2023. "Regulation of the Epithelial to Mesenchymal Transition in Osteosarcoma" Biomolecules 13, no. 2: 398. https://doi.org/10.3390/biom13020398

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