Next Article in Journal
The Plant Fatty Acyl Reductases
Next Article in Special Issue
Cellular and Molecular Biology of Cancer Stem Cells of Hepatocellular Carcinoma
Previous Article in Journal
Detection of the Omicron SARS-CoV-2 Lineage and Its BA.1 Variant with Multiplex RT-qPCR
Previous Article in Special Issue
Bioinformatics Analysis of RNA-seq Data Reveals Genes Related to Cancer Stem Cells in Colorectal Cancerogenesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Tumor Microenvironment in Regulating the Plasticity of Osteosarcoma Cells

MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(24), 16155; https://doi.org/10.3390/ijms232416155
Submission received: 10 November 2022 / Revised: 7 December 2022 / Accepted: 15 December 2022 / Published: 18 December 2022
(This article belongs to the Special Issue Cellular and Molecular Biology of Cancer Stem Cells)

Abstract

:
Osteosarcoma (OS) is a malignancy that is becoming increasingly common in adolescents. OS stem cells (OSCs) form a dynamic subset of OS cells that are responsible for malignant progression and chemoradiotherapy resistance. The unique properties of OSCs, including self-renewal, multilineage differentiation and metastatic potential, 149 depend closely on their tumor microenvironment. In recent years, the likelihood of its dynamic plasticity has been extensively studied. Importantly, the tumor microenvironment appears to act as the main regulatory component of OS cell plasticity. For these reasons aforementioned, novel strategies for OS treatment focusing on modulating OS cell plasticity and the possibility of modulating the composition of the tumor microenvironment are currently being explored. In this paper, we review recent studies describing the phenomenon of OSCs and factors known to influence phenotypic plasticity. The microenvironment, which can regulate OSC plasticity, has great potential for clinical exploitation and provides different perspectives for drug and treatment design for OS.

1. Introduction

Osteosarcoma (OS) is a malignancy that most commonly occurs in children and adolescents and is the second highest cause of cancer-related mortality in these groups [1,2,3]. There has been a rise in the annual incidence rate of OS to three cases per million individuals [4]. The majority of OS cases arises in the metaphyseal regions adjacent to the physis, including the distal femur, proximal tibia and the proximal humerus, with a strong capacity for proliferation [5]. Over the past 30 years, the treatment of OS has improved little, such that surgery accompanied with chemoradiotherapy remain as the main method of treatment [6]. Although novel clinical strategies such as gene editing, individualized treatment and novel molecular-targeted therapies, e.g., angiogenesis inhibitors, tyrosine kinase inhibitors and monoclonal antibodies, have all been deployed against OS, the outcomes for patients are poor, particularly those with more aggressive forms of the cancers [3,4,5,6,7]. Therefore, novel treatment strategies are in demand in clinical practice. In addition, the molecular mechanism underlying tumorigenesis and malignant metastasis needs to be studied in detail.
Based on the present research, OS is speculated to have two main origins, bone mesenchymal stem cells (BMSCs) and osteoblasts [8,9,10]. p53 as a classic cancer suppressor gene plays a key role in OS progression. The deficiency of p53 is an important reason leading to primary OS. In addition, retinoblastoma gene (Rb), cyclin dependent kinase inhibitor 2 (CDKN2), KRAS and c-Met also participate in the regulation of OS progression [8,9,10,11]. Within cancer tissues, there exist several dynamic subsets of cancer cells considered to be cancer stem cells (CSCs) or stem cell-like cancer cells [12,13]. CSCs have been frequently reported to exhibit stem cell properties and capabilities of long-term clonal proliferation, tumorigenicity, facilitating metastasis and promoting resistance to chemotherapy and radiotherapy [14,15]. Therefore, exploring the origins of cancer initiation and metastasis will likely facilitate the development of future therapies. In 1994, Lapidot et.al first reported that, in human acute myeloid leukemia, a rare population of CSCs exists [16]. Subsequently, an accumulating number of studies have also reported the existence of CSCs in other solid tumors, including prostate, glioblastoma, hepatoma, breast cancer and OS [17,18,19,20,21,22]. In fact, all types of malignant tumors consist of different subpopulations of tumor cells, leading to high degrees of heterogeneity.
The niche in which CSCs reside is the tumor microenvironment, where they co-exist with adjacent supporting cells, micro-vessels and the extracellular matrix [19,20]. In addition, the tumor microenvironment can contain soluble factors, such as chemokines and cytokines, whilst being under the influence of various mechanical factors, including matrix stiffness, solid stress and fluid stress [23,24]. In the OS microenvironment, OS stem cells (OSCs) are contained in a specialized niche that contains a unique bone microenvironment, which consists of various types of bone cells, such as osteoblasts or osteoclasts. OSCs are similar to other CSCs, in that they account for a proportion of cancer cells with tumorigenic and self-renewal capabilities. The existence of OSCs was first confirmed by Gibbs et al., who found that when primary human OS cells or the OS cell line MG63 were suspended in a serum-free medium with defined growth factors, 0.1% of the cells could form spheres with self-renewal capacity [17,18,19,20,21,22,23,24,25]. Subsequently, a series of studies have proven the existence of OSCs, in addition to revealing the phenotype and possible marker profile of OSCs. Here, we summarize and review the recent studies on possible OSC markers and phenotypes (Table 1).
The primal CSC theory is a hierarchical model that proposes that CSCs have similar patterns compared with normal stem cells. According to this theory, CSCs can either undergo asymmetric fission, where they can divide to give two different progenies (one CSC and the other non-CSC), or symmetrically into two CSCs [43]. CSCs have shown great potential as a target for cancer chemotherapy according to this hierarchical model. Stem cell research strategies have been previously applied for the exploration of CSCs, specifically of the specific markers of CSCs [44,45]. However, thus far it has not been possible to identify a marker that can be used to definitively identify cancer cells with ‘stem’ characteristics in distinct tumors and tumor cell lines. To date, there is no special marker that can accurately represent or identify CSCs in any cancer type or cell line. Previous studies have even suggested that mature leukemic progenitor cells or cells expressing lineage markers can initialize tumorigenesis [46,47]. This hierarchic model has been questioned. Accordingly, a new stochastic theory of tumorigenesis has been proposed. The new theory states that each cancer cells can switch phenotypes to gain the CSC phenotype under certain conditions in the microenvironment. According to this theory, non-CSCs can transform into CSCs if certain parameters in the tumor microenvironment are met [48,49,50]. Indeed, a series of studies have previously demonstrated that, under certain conditions or after eradication of CSCs, some non-CSCs can transform to gain the CSC phenotype to facilitate tumor progression [51,52,53].
Importantly, since tumors are comprised of various different cell types and not only cancer cells, but this inter-conversion does also not only affect CSCs and non-OSCs. In fact, the tumor microenvironment is highly complex and can contain other cell types, such as immune cells and cancer-associated fibroblasts. It can also contain soluble factors and mechanical components, such as growth factors. In addition, the tumor microenvironment can come under the influence of various stimuli, such as hypoxia. In this review, we summarize the research describing the phenomenon of cancer cell reversion, especially for OS. Furthermore, the factors that have been shown to influence cell plasticity switch are mentioned. The mechanism underlying this reversion, the influence of the tumor microenvironment composition and their overall effect on the metastatic spread of the disease are also discussed.

2. Role of the Tumor Microenvironment in Regulating OSC Stemness

OSCs can interact with their microenvironment through complex and dynamic processes, including variation of oxygen, mechanical interactions, enzymatic modification of the extracellular matrix (ECM) structure and signaling cross-talk, all of which can influence the progression and the dissemination of OS cells (Figure 1)

2.1. Hypoxia

Common hallmarks of solid tumors include intra-tumoral hypoxia, necrosis, acidic environments and disturbed angiogenesis. Previous studies have shown that increased cancer cell stemness is associated with intra-tumoral hypoxia [54,55,56,57]. Our previous study demonstrated that a hypoxia microenvironment could induce non-OSCs dedifferentiation into OSCs by increasing the expression of TGF-β [58].
During the process of tumor formation, excessive proliferation of cancer cells consumes large quantities of oxygen in the microenvironment, resulting in the formation of a hypoxic zone in the central area of the tumor. In addition, aberrant secretion of angiogenetic factors, including vascular endothelial growth factor A and fibroblast growth factor 2 (FGF2), result in malformation and disorder in the neovascularization system [54,55]. This in turn causes the loss of oxygen supply, further aggravating hypoxia in the cancer tissue [59,60].
This hypoxic microenvironment induces the expression of hypoxia-inducible factor-1 (HIF-1), a vital member of the HIF family. By contrast, in the presence of oxygen, HIF-1 undergoes degradation by the von Hippel-Lindau protein, a tumor suppressor protein [61,62,63,64,65]. HIF-1 is a heterodimer that is ubiquitously expressed in human and mouse tissues. HIF-1 consists of two subunits, the hypoxia-inducible, oxygen-dependent subunit HIF-1α and the constitutively-expressed oxygen-independent subunit HIF-1β [66,67,68]. It is only when the oxygen concentration reaches <5% (such as when the volume of the tumor has grown to >300 mm2) that HIF-1α can exist stably. Activity of HIF-1 provides cancer cells with the ability to adapt to hypoxia and is closely associated with tumor metabolism, differentiation, angiogenesis, cell proliferation, metastasis and multidrug resistance. Of note, several studies have demonstrated that elevated expression of HIF-1α promoted the dedifferentiation of cancer cells into CSCs, whereas hypoxia is directly associated with poorer prognoses in patients with OS [69].
Numerous studies have demonstrated that hypoxia can promote the expression of the stem cell marker CD133 to maintain stemness and drug resistance in the Saos-2 OS cell line [70]. Lin et al. previously reported that hypoxia can increase the expression of embryonic stem cell markers, including Oct3/4 and Nanog, in the MNNG/HOS OS cell line [71]. Zhang et al. showed that a hypoxic microenvironment stabilized HIF-1α in OS cells, such that HIF-1 promoted the expression of microRNA (miR or miRNA)-210, which then induced and accelerated the dedifferentiation of OS cells into OSCs [58,59,60,61,62,63,64,65,66,67,68,69,70,71,72]. These observations aforementioned suggest that HIF-1 and subsequent hypoxia signaling pathways can regulate the differentiation of CSCs and the dedifferentiation of non-stem cells in tumors [73,74].
In addition to HIF, hypoxia can also cause integrin-linked kinase dysfunction, triggering CSCs formation [75]. Hypoxia has been previously found to promote breast cancer stemness by HIF-dependent and AlkB homolog 5-mediated N6-methyladenosine (m6A)-demethylation of Nanog mRNA [76]. Shi et al. used the evolutionary theory to identify the hypoxic adaptation-associated gene YTH N6-methyladenosine RNA binding protein 1 (YTHDF1). As a member of the N6-methyladenosine (m6A)-modified RNA-binding protein family, YTHDF1 may interplay with other m6A modifiers and serve a pivotal role in the self-renewal and differentiation of stem cells [77]. Under hypoxia, AKT will accumulate in the mitochondria of tumor cells, whereby 3-phosphoinositide-dependent protein kinase 1 is phosphorylated at special sites. This pathway shifts the tumor metabolic program to glycolysis, which antagonizes apoptosis and autophagy and inhibits oxidative stress. This in turn maintains the survival and proliferation capabilities of tumor cells, as evidenced by the sphere-forming ability of cells in 3D cultures under severe hypoxia [78].
These aforementioned findings suggest that hypoxia may contribute to the creation of a microenvironment rich in tumor stem cells, where this unique hypoxic microenvironment may provide essential cellular interactions and environmental signals for the maintenance of CSCs [70,71,72,73,74,75,76,77,78,79]. By contrast, the hypoxia microenvironment can also regulate non-CSC dedifferentiation by regulating the activities of other pathways, including epithelial-mesenchymal transition (EMT), metabolic reprogramming, DNA hypermethylation and apoptotic resistance. Additionally, the hypoxic microenvironment can mediate the resistance of CSCs against drugs through drug transporters [80]. The majority of CSCs express the ATP-binding cassette (ABC) family of membrane transporters at high levels, including multidrug resistance gene 1, breast cancer resistance protein and multidrug resistance-associated protein. These proteins can transport metabolites, drugs and other substances, allowing CSCs to become highly resistant to chemotherapy. The relationship between hypoxia and ABC proteins was previously documented to have a strong association with mediating tumor drug resistance [81].
In conclusion, the hypoxic microenvironment with the activation of hypoxic signaling can serve key roles in the dedifferentiation of OS cells into OSCs. Therefore, it is important to study the molecular mechanism underlying OS dedifferentiation, which is expected to hold important clinical significance for improving the efficacy of therapeutic strategies. However, the molecular mechanism of how exactly the hypoxic microenvironment can regulate OSC physiology biology requires additional experimental evidence for validation. In particular, HIF-1 is a key molecule of the hypoxia signaling pathway, the downstream molecules of which are expected to become important markers and potential molecular targets of OSCs.

2.2. Biomechanical Force

Under physiological conditions, most if not all organisms experience complex biomechanical forces, including shear stress, matrix stiffness, tension and compression pressure [82,83,84,85,86]. Biomechanical forces experienced by solid tumors have different profiles compare with those in the surrounding or healthy tissue [87]. Throughout the process of cancer development, excessive cell proliferation will lead to the abnormal development of the biomechanical microenvironment, including solid stress, increased matrix stiffness (decrease in OS due to osteolysis) and abnormal interstitial fluid pressure [88,89,90].
These complex mechanical systems are essential for the maintenance of the homeostasis of the CSC population. Indeed, previous studies have demonstrated that non-CSCs can be transformed into CSCs by receiving mechanical signals from the surrounding microenvironment, such as increased matrix stiffness [91,92,93,94] and/or fluid shear stress [95,96,97,98]. In OS, soft substrate (7 kPa) has been reported to preserve OS stemness, mainly through miR-29b/Spin 1-dependent signaling. Manipulation of cancer niche stiffness and miR-29b expression may therefore be potentially novel drug targets in OS [99]. Previous studies have shown that EMT can promote the progression and invasion of tumors [100]. EMT have been observed to serve as a direct link between non-CSCs and the gain of CSC properties [101]. Matrix stiffness in the tumor microenvironment can actively regulate EMT and migration of OS cells through cytoskeletal remodeling and the translocation of myocardin related transcription factor A, which may contribute to cancer progression [102]. Although the aforementioned studies revealed that mechanical factors are at least partially associated with the dynamic conversion between non-CSCs and CSCs, further research into the association between mechanical signaling and OSC stemness is warranted.
Mechanical receptors on the cell surface, such as integrins, CD44 and ion channels, can sense the changes in ECM and activate key downstream molecules, including focal adhesion kinase, integrin-linked kinase, RhoA and yes-associated protein. Several of the signals induce non-CSC reprogramming and transform them into CSCs by increasing the expression of sex determining region Y-box 2 (Sox2), octamer-binding transcription factor (Oct)-4 and Nanog [91,92,93,94,95,96,97,98,99,100,101,102,103,104]. CSCs and normal stem cells frequently share similar surface markers and signaling pathways, which would restrict the design of treatment regimens [105]. The abnormal mechanical system in OS microenvironments, which rarely occur in the harmonious microenvironments of normal stem cells, may provide novel insights for designing CSC-targeted treatment methods. As such, discovering the relationship between biomechanical factors and CSCs will greatly enable the generation of novel research strategies to investigate the occurrence, development, and recurrence of cancers.

2.3. Growth Factors

Growth factors are pivotal in maintaining the physiological behavior of healthy individuals. Cells in the tumor microenvironment can secrete growth factors to regulate processes of tumor development [106,107]. When OS arise in the bone, OS cells secrete factors that direct osteoclast-mediated bone destruction. In addition, matrix-derived growth factors, especially transforming growth factor β1 (TGF-β1), are released from bone matrix. In addition, OS cells can release TGF-β1 directly, where increased TGF-β1 expression is associated with high-grade metastases of OS [108]. TGF-β1 is a multi-function cytokine that serves as a mediator in the tumor to facilitate further tumor expansion, metastasis and cytokine production [109]. Wang et.al previously reported that TGF-β1 can switch the OSC chemoresistance through the miR-499a/SHKBP1 axis [110]. In another study, TGF-β1 signaling and a hypoxic environment were found to induce the transformation of non-OSCs into OSCs dynamically, which promoted the acquisition of chemoresistance, tumorigenicity, neovasculo-genicity and metastatic potential. Furthermore, blocking the TGF-β1 signaling pathway was reported to inhibit this switch from non-OSCs to OSCs, inhibit OSC self-renewal and suppress hypoxia-mediated dedifferentiation [58]. In the bone microenvironment, TGF-β1 signaling is responsible for OSC generation and critical to chemoresistance in vivo. In addition to OS, TGF-β1 can also regulate the dynamic switching between stem cells and non-stem cells to influence the progression of tumors from different tissue origins [111,112]. In conclusion, TGF-β1 serves a key role in regulating the dynamic plasticity of OSC, which can lead to non-stem cells adopting OSC characteristics to promote tumorigenesis and chemoresistance, highlighting TGF-β1 as a potential therapeutic target.
Bone morphogenetic proteins (BMPs) are members of the TGF-β superfamily and serve important roles in the activity of various tissues. In OS, BMP-2 suppresses tumor growth by reducing the expression of oncogenes whilst promoting the differentiation of OSCs [113]. Histological examination and gene expression analysis of OS tissues revealed that fibrotic remodeling of the tumor microenvironment favors tumorigenesis. Zhang et al. previously demonstrated that fibrotic reprogramming in the lung induced by OSCs is critical for OS pulmonary metastasis, with FGF-FGF receptor 2 (FGFR2) signaling being responsible for this important process [114]. In OS, the tumor necrosis factor-α/miR-155 axis has been found to induce OSC transformation between non-OSCs and OSCs through the extracellular signal-regulated protein kinase signaling pathway [115]. Melatonin, one of the hormones secreted by the pineal gland of the brain, has been shown to significantly inhibit sphere formation by OSCs through the key transcription factor Sox-9 [116]. Although all of the aforementioned growth factors have shown the potential to target OSCs, the underlying mechanism require further exploration.

2.4. Cancer-Associated Cells

Together with intrinsic tumor cell changes, other cell types in the surrounding microenvironment, including fibroblasts, endothelial cells, immune cells and mesenchymal stromal cells (MSCs), can all contribute functional variety by interacting with the tumor cells [117]. MSCs within the microenvironment have been known to secrete a number of cytokines. It has been frequently reported that cytokine production by tumor-associated stroma can stimulate tumor sustenance, growth and angiogenesis, where in OS this is no exception. In addition, several reports have previously found that MSCs can produce soluble factors to regulate cancer cell stemness [118]. In particular, MSCs can secrete TGF-β1 and interleukin (IL)-6, which in turn increases stemness, cell proliferation, migration and the metastatic potential of OSCs [115]. Towards OSCs, MSCs can also increase the expression of adhesion molecules, such as intercellular adhesion molecule-1 [119].
The immune component of the OS microenvironment is mainly comprised of tumor-associated macrophages (TAMs), myeloid-derived suppressor cells, dendritic cells and regulatory T cells. Macrophages can engulf and digest foreign substances to clear potentially harmful material, such as tumor cells [120]. M2 macrophages were found to be enriched in OS tissues. M2-type TAMs have been reported to promote OS cell stemness by upregulating the expression of stemness markers whilst facilitating colony formation, sphere formation and tumor initiation [121]. All-trans retinoic acid treatment has been documented to prevent the M2 polarization of TAMs, which then diminished the CSC phenotypes, including colony- and sphere-forming capabilities [121]. TAMs have also been shown to reinforce the CSC populations through direct interactions between ephrin and ephrin type A receptor 4. This leads to the production of inflammatory cytokines IL-1, IL-6 and IL-8 by CSCs, which sustains the CSC state [122,123]. However, the mechanism by which TAMs can upregulate the CSC-like phenotype in OS remains unknown.
It is becoming accepted that fibrotic remodeling of the tumor microenvironment generally favors tumorigenesis. Myofibroblasts synthesize and deposit matrix fibrils into the extracellular space, which is one of the hallmarks of fibrosis. A previous study demonstrated that inducing fibrotic reprogramming in OSCs is critical for the growth of lung metastases. Fibronectin auto-deposition has been observed to sustain fibro-genic reprogramming and OSC proliferation, which resembles the process that occurs when non-malignant myofibroblasts induce tissue fibrosis [114].
Complex cell-to-cell interactions are necessary for maintaining the homeostasis of the CSC population. All cellular components in the tumor niche, including CSCs, non-CSCs, fibroblasts, immune cells and mesenchymal cells, can perceive the modulator effects and soluble proteins emitted by CSCs, resulting in non-CSC and CSCs interconversion. In turn, other cell types in the tumor microenvironment activate paracrine signaling pathways to ensure CSC proliferation and dissemination. However, there remains numerous as yet unknown mechanisms mediated by tumor-associated cells in the microenvironment that can regulate the plastic transition between non-CSCs and OSCs.

3. Extracellular Vesicles (EVs)

EVs are small membranous vesicles released by cells into the extracellular matrix. EVs are abundant throughout in the body and can stably carry and transfer important signaling molecules between cells, serving as another mechanism of cell–cell communication [124,125]. Accumulating evidence has shown that EVs may also be important in regulating OS development, progression and metastasis, supporting the notion that EVs can be of use as potential biomarkers for the diagnosis and prognosis of OS [126,127,128,129].
Previous studies have reported the potential biological role of exosomes in CSCs. Yang et.al found that human umbilical vein endothelial cell (HUVEC)-derived exosomes enhanced the proportion of CD117+ cells relative stemness gene, which increased sphere formation. Furthermore, HUVEC-exosomes have been found to promote cell stemness in OS by activating the Notch signaling pathway [130]. Zhang et.al demonstrated that bone mesenchymal stem cell (BMSC)-derived EVs could be transferred into OS cells to inhibit tumor progression by targeting transformer 2β homolog (TRA2B), before subsequently proposing the potential of miR-206 and TRA2B as novel therapeutic targets [131]. In another previous study, BMSC-derived EVs were found to activate the miR-30-5p/Kruppel-like factor 10 axis in OS cells to promoting cell proliferation and lung metastasis [132].
In pancreatic cancer, EVs have been observed to exert significant effects on regulating CSCs. M2 macrophage-derived EVs have been documented to promote pancreatic cancer stem cell differentiation and activities through miR-21-5p [133]. EVs derived from chemo-sensitive non-stem bladder cancer cells were found to be enriched with cargo proteins that can mediate proteo-static functions to significantly alter the development of CSCs [130]. As a result, they became more intrinsically chemo-resistant and aggressive with enhanced self-renewal capabilities [134]. Furthermore, EVs derived from human liver stem cells or MSCs have been found to reduce CSC proliferation and invasion whilst increasing CSC apoptosis, but had no effect on non-CSCs [135].
Due to their biogenesis, EVs may contain a high variety of molecular cargoes that is dependent on their cell of origin. DNA, proteins, mRNA, microRNA and lipids, all of which can regulate signaling pathways inside target cells, have been documented [136]. There is ample evidence that CSC-derived EVs can promote stem-like properties in non-CSCs, leading to the enhanced tumorigenicity [137,138,139]. For these reasons, EVs are now considered to be leading facilitators in promoting the dynamic interconversion between non-CSCs and CSCs. However, in OS, experimental research data on the composition of EVs in OSCs remaining lacking at present. In addition, the components within EVs secreted by different cell types in the OS microenvironment remain obscure. Further research is required to characterize the cellular and noncellular components of the OS microenvironment to understand how it regulates OSC plasticity.

4. Non-Coding RNAs (ncRNAs) in Regulating OSCs

Developmental signaling pathways, including the Notch, Wnt, Hedgehog and Hippo pathways, are commonly found to be dysregulated in CSCs. Indeed, these signaling pathways can all serve key regulatory functions that support the maintenance and survival of CSCs. Further research is needed to deepen the understanding into these signaling pathways aforementioned in OSCs. In recent years, research into the roles of non-coding RNAs in OSCs has been on the increase. A series of studies have previously shown that (ncRNAs) can regulate the dynamic OSC plasticity (Figure 2) [140]. ncRNAs are RNA sequences that do not encode proteins but can regulate gene expression, which include micro miRNAs (miRNAs), long ncRNAs (lncRNAs) and circular RNAs (circRNAs) [141]. miRNAs are typically 18–25 nucleotides in length and target specific mRNAs by either complete or partial complementary binding to their 3′untranslated regions (UTR) [142]. By contrast, lncRNAs are >200 nucleotides in length and mainly exert their functions by sponging miRNAs and targeting specific substrates [143].

4.1. miRNAs

In recent years, miRNAs have been reported to regulate CSCs in a variety of cancers, including OS [144]. Research in our laboratory has shown that miR-34a serve a key role in regulating non-OSC dedifferentiation into OSCs. The miR-34 could inhibit OS dedifferentiation into OSCs through the Sox2-PAI pathway [145]. miR-26a expression has been found to be reduced in OSCs, such that miR-26a overexpression was able to inhibit sphere formation and tumor cell proliferation both in vitro and in vivo. miR-26a can also inhibit OS cell stemness by targeting Jagged1 expression, one of the Notch ligands [146]. In another study, Zou et.al previously revealed that miR-34a expression was lower in OSCs, where overexpression of miR-34a reduced sphere formation ability and the expression stem cell marker genes [147]. Liang et.al also suggested that increased miR-34a expression in OS can inhibit sphere and colony formation abilities [148]. In addition, Zhao et.al reported that miR-1247 can inhibit CD117+ CSC sphere formation and stem cell-associated gene expression [149]. Patients with OS and lower expression levels of miR-382 were associated with poorer chemotherapy responses and poorer prognoses, whilst upregulating the expression of miR-382 was found to inhibit pulmonary metastasis and reduced the population of OSCs [150]. Increasing miR-29b-1 expression was also found to increase drug susceptibility in OS cells. In addition, miR-29b-1 overexpression could also inhibit the expression of stem cell genes by suppressing Oct3/4, Sox2 and Nanog in OSCs [151]. In another previous study, Guo et.al reported that OSCs have lower miR-335 expression levels, because miR-335 inhibited OS cell stemness by suppressing the Oct-4 pathway [152]. Overexpression of the miRNA lethal-7 has been reported to reduce sphere formation and decrease the expression of stem cell markers in OSCs.

4.2. lncRNAs

Recently, a number of studies have reported that lncRNA can alter the balance between non-OSCs and OSCs. One previous study found that expression of the lncRNA differentiation antagonizing non-protein coding RNA (DANCR) was increased in OS tissues and OS cell lines. DANCR can also enhance tumor malignancy by increasing the OSC pool through activation of the PI3K/AKT signaling pathway in OS [153]. LncRNA DLX6-antisense 1 (AS1) overexpression in OS was reported to promote stemness in OS cells through miR-129-5p activating Wnt pathway. Zhang et.al. also found that higher expression levels of DLX6-AS1 in OS tissues tended to associate with poorer prognosis [154]. In addition, overexpression of LncRNA hypoxia-inducible factor-2α promoter upstream transcript decreased the population of CD133+ OSCs, which inhibited the sphere-forming capacity [155]. LncRNA testis-associated oncogenic lncRNA was shown to directly bind to the 3′-UTR of SOX9 mRNA to increase stemness in the OS cell line MG-63 [156]. Ma et.al. revealed that inhibition of lncRNA fer-1 family member 4 upregulated the expression of stemness related marker [157]. LncRNA long intergenic non-coding RNA for kinase activation was found to at least in part govern the stemness of OS by decreasing the percentage of CD133+ cells in OS cell lines [158]. Chen et.al. reported that lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is highly expressed in tumor tissues and is associated with tumor size, metastasis and poorer survival in patients with OS. Furthermore, MALAT1 has been discovered to regulate stem cell expression in OS [159]. LncRNA SOX2 overlapping transcript (OT) was found to be overexpressed in OS cell lines and patients with OS, where knocking down the expression of this lncRNA could significantly decrease the expression of stemness biomarker [160]. Li et.al. demonstrated that lncRNA β-1,4-galactosyltransferase 1-AS1 knockdown inhibited sphere formation and decreased stemness marker expression [161]. Shi et.al. reported that circRNA phosphatidylinositol-4-phosphate 5-kinase type 1α could significantly inhibit sphere formation by OS cells and decreased the CD133 + /CD44+ cell population [162].

5. Targeting the Microenvironment of OSCs

The complex interactions among various components and tumor cells within a specific microenvironment serves a critical role in tumor growth and dissemination. Cancer cells require mechanical support, blood supply and growth factors secreted by cancer associated cells to guarantee continuous growth [163]. The crosstalk between OSCs and the microenvironment provides a variety of potential therapeutic strategies for the targeted treatment of OSCs, in theory. However, OSCs can resist apoptosis using various methods [164], allowing them to survive the clinical treatments currently available [10]. In addition, a number of treatment strategies may even induce non-OSC transformation into CSCs [165].
TGF-β1 as a key component of the tumor microenvironment and can regulate the interconversion between CSC and non-CSCs, suggesting that it has potential as a treatment target. A strategy combining a nanoparticle-based vaccine with the targeted silencing of TGF-β expression using liposome-protamine-hyaluronic acid nanoparticles has been previously attempted. Since TGF-β can function as an immune suppressor, silencing it may enhance the systemic immune response against the tumor [166]. Fresolimumab (GC-1008) as TGF-β inhibitor is used to treat CSCs and GC-1008 IN PhaseⅠorⅡ, correspondingly for malignant melanoma or metastatic breast cancer [167]. Besides, inducing CSC differentiation is also an efficient strategy and a series of drugs aiming Wnt or Notch signal pathway also have great therapy potential [168,169]. Notch signaling constitutes a highly conserved cell fate determining pathway with functions pertinent to a wide breadth of cancer biology, including the CSC phenotype, angiogenesis, metastasis and tumors immune evasion. A new drug, rovalpituzumab tesirine (Rova-T) conjugated DDL3 (an atypical Notch ligand) antibody, is performing in Phase Ⅲ for small-cell lung cancer [170]. Salinomycin can selectively target OSCs to inhibit OS cell proliferation through the Wnt/β-catenin signaling pathway [171]. In addition, other attractive CSC immunotherapeutic targets supported by preclinical data include chemokine receptors, such as the IL-8 receptor C-X-C motif chemokine receptor 1, IL-8 and IL-6 [172]. Furthermore, IL-6 and IL-8 inhibitors tocilizumab and reparixin have been demonstrated to show potential in chemotherapy.
Although targeting OSCs is considered to be one of the more promising research fields as a novel treatment strategy, the majority of OSC-based treatments have not been able to successfully enter clinical trials. A variety of reasons have been proposed for this suboptimal translation from bench to bedside, such as inadequate physicochemical features of OSCs and scarce knowledge of the interconversion mechanism.

6. Conclusions

According to the dynamical OS cell plasticity, the most promising strategies for preventing OS metastasis should be those that can target the activator of the OSCs instead of the OS itself. This is because of the potentially dynamic swing between OSCs and non-OSCs. Deciphering the OS microenvironment would lead to a deeper understanding of the fundamental nature of the OSC/non-OSC crosstalk, the interconversion between non-OSCs and OSCs and ultimately their impact on future clinical treatment outcomes. Likewise, exploring OS cell reprogramming occurring in the TME should also open the possibility of designing novel strategies to combat OS relapse and metastatic spread.

Author Contributions

B.T.: Conceptualization, Writing—Original Draft, Writing—Review & Editing; X.D.: Writing—Original Draft; S.Z.: Visualization; Y.Z.: Writing—Review & Editing, Supervision, Project administration, Funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

The present project was supported by a grant from the National Natural Science Foundation of China (No. 31871413) and two from the Programs of Guangdong Science and Technology (2017B020230002 and 2016B030231001).

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. Turcotte, L.M.; Neglia, J.P.; Reulen, R.C.; Ronckers, C.M.; van Leeuwen, F.E.; Morton, L.M.; Hodgson, D.C.; Yasui, Y.; Oeffinger, K.C.; Henderson, T.O. Risk, Risk Factors, and Surveillance of Subsequent Malignant Neoplasms in Survivors of Childhood Cancer: A Review. J. Clin. Oncol. 2018, 36, 2145–2152. [Google Scholar] [CrossRef] [PubMed]
  2. Grünewald, T.G.P.; Cidre-Aranaz, F.; Surdez, D.; Tomazou, E.M.; de Álava, E.; Kovar, H.; Sorensen, P.H.; Delattre, O.; Dirksen, U. Ewing sarcoma. Nat. Rev. Dis. Prim. 2018, 4, 5. [Google Scholar] [CrossRef] [PubMed]
  3. Smith, R.A.; Andrews, K.S.; Brooks, D.; Fedewa, S.A.; Manassaram-Baptiste, D.; Saslow, D.; Wender, R.C. Cancer screening in the United States, 2019: A review of current American Cancer Society guidelines and current issues in cancer screening. CA A Cancer J. Clin. 2019, 69, 184–210. [Google Scholar] [CrossRef] [PubMed]
  4. Gianferante, D.M.; Mirabello, L.; Savage, S.A. Germline and somatic genetics of osteosarcoma—Connecting aetiology, biology and therapy. Nat. Rev. Endocrinol. 2017, 13, 480–491. [Google Scholar] [CrossRef] [PubMed]
  5. Bielack, S.S.; Kempf-Bielack, B.; Delling, G.; Exner, G.U.; Flege, S.; Helmke, K.; Kotz, R.; Salzer-Kuntschik, M.; Werner, M.; Winkelmann, W.; et al. Prognostic factors in high-grade osteosarcoma of the extremities or trunk: An analysis of 1,702 patients treated on neoadjuvant cooperative osteosarcoma study group protocols. J. Clin. Oncol. 2002, 20, 776–790. [Google Scholar] [CrossRef]
  6. Crompton, J.G.; Ogura, K.; Bernthal, N.M.; Kawai, A.; Eilber, F.C. Local Control of Soft Tissue and Bone Sarcomas. J. Clin. Oncol. 2018, 36, 111–117. [Google Scholar] [CrossRef] [Green Version]
  7. Buja, A.; Lago, L.; Lago, S.; Vinelli, A.; Zanardo, C.; Baldo, V. Marital status and stage of cancer at diagnosis: A systematic review. Eur. J. Cancer Care 2018, 27, e12755. [Google Scholar] [CrossRef]
  8. Rubio, R.; Abarrategi, A.; Garcia-Castro, J.; Martinez-Cruzado, L.; Suarez, C.; Tornin, J.; Santos, L.; Astudillo, A.; Colmenero, I.; Mulero, F.; et al. Bone environment is essential for osteosarcoma development from transformed mesenchymal stem cells. Stem Cells 2014, 32, 1136–1148. [Google Scholar] [CrossRef]
  9. Mohseny, A.B.; Szuhai, K.; Romeo, S.; Buddingh, E.P.; Briaire-de Bruijn, I.; de Jong, D.; van Pel, M.; Cleton-Jansen, A.M.; Hogendoorn, P.C. Osteosarcoma originates from mesenchymal stem cells in consequence of aneuploidization and genomic loss of Cdkn2. J. Pathol. 2009, 219, 294–305. [Google Scholar] [CrossRef]
  10. Basu-Roy, U.; Basilico, C.; Mansukhani, A. Perspectives on cancer stem cells in osteosarcoma. Cancer Lett. 2013, 338, 158–167. [Google Scholar] [CrossRef]
  11. Zhang, Y.; Mai, Q.; Zhang, X.; Xie, C.; Zhang, Y. Microenvironment Signals and Mechanisms in the Regulation of Osteosarcoma. Osteosarcoma Biol. Behav. Mech. 2017. [Google Scholar] [CrossRef]
  12. Teng, Y.D.; Wang, L.; Kabatas, S.; Ulrich, H.; Zafonte, R.D. Cancer Stem Cells or Tumor Survival Cells? Stem Cells Dev. 2018, 27, 1466–1478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Medema, J.P. Cancer stem cells: The challenges ahead. Nat. Cell Biol. 2013, 15, 338–344. [Google Scholar] [CrossRef] [PubMed]
  14. Arima, Y.; Nobusue, H.; Saya, H. Targeting of cancer stem cells by differentiation therapy. Cancer Sci. 2020, 111, 2689–2695. [Google Scholar] [CrossRef]
  15. Batlle, E.; Clevers, H. Cancer stem cells revisited. Nat. Med. 2017, 23, 1124–1134. [Google Scholar] [CrossRef] [PubMed]
  16. Lapidot, T.; Sirard, C.; Vormoor, J.; Murdoch, B.; Hoang, T.; Caceres-Cortes, J.; Minden, M.; Paterson, B.; Caligiuri, M.A.; Dick, J.E. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994, 367, 645–648. [Google Scholar] [CrossRef]
  17. Gibbs, C.P.; Kukekov, V.G.; Reith, J.D.; Tchigrinova, O.; Suslov, O.N.; Scott, E.W.; Ghivizzani, S.C.; Ignatova, T.N.; Steindler, D.A. Stem-like cells in bone sarcomas: Implications for tumorigenesis. Neoplasia 2005, 7, 967–976. [Google Scholar] [CrossRef] [Green Version]
  18. Klarmann, G.J.; Hurt, E.M.; Mathews, L.A.; Zhang, X.; Duhagon, M.A.; Mistree, T.; Thomas, S.B.; Farrar, W.L. Invasive prostate cancer cells are tumor initiating cells that have a stem cell-like genomic signature. Clin. Exp. Metastasis 2009, 26, 433–446. [Google Scholar] [CrossRef]
  19. Yuan, X.; Curtin, J.; Xiong, Y.; Liu, G.; Waschsmann-Hogiu, S.; Farkas, D.L.; Black, K.L.; Yu, J.S. Isolation of cancer stem cells from adult glioblastoma multiforme. Oncogene 2004, 23, 9392–9400. [Google Scholar] [CrossRef] [Green Version]
  20. Cao, L.; Zhou, Y.; Zhai, B.; Liao, J.; Xu, W.; Zhang, R.; Li, J.; Zhang, Y.; Chen, L.; Qian, H.; et al. Sphere-forming cell subpopulations with cancer stem cell properties in human hepatoma cell lines. BMC Gastroenterol. 2011, 11, 71. [Google Scholar] [CrossRef]
  21. Ma, S.; Chan, K.W.; Hu, L.; Lee, T.K.; Wo, J.Y.; Ng, I.O.; Zheng, B.J.; Guan, X.Y. Identification and characterization of tumorigenic liver cancer stem/progenitor cells. Gastroenterology 2007, 132, 2542–2556. [Google Scholar] [CrossRef]
  22. Ponti, D.; Costa, A.; Zaffaroni, N.; Pratesi, G.; Petrangolini, G.; Coradini, D.; Pilotti, S.; Pierotti, M.A.; Daidone, M.G. Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/progenitor cell properties. Cancer Res. 2005, 65, 5506–5511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Plaks, V.; Kong, N.; Werb, Z. The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 2015, 16, 225–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ju, F.; Atyah, M.M.; Horstmann, N.; Gul, S.; Vago, R.; Bruns, C.J.; Zhao, Y.; Dong, Q.Z.; Ren, N. Characteristics of the cancer stem cell niche and therapeutic strategies. Stem Cell Res. Ther. 2022, 13, 233. [Google Scholar] [CrossRef]
  25. Gibbs, C.P., Jr.; Levings, P.P.; Ghivizzani, S.C. Evidence for the osteosarcoma stem cell. Curr. Orthop. Pract. 2011, 22, 322–326. [Google Scholar] [CrossRef] [PubMed]
  26. Tirino, V.; Desiderio, V.; d’Aquino, R.; De Francesco, F.; Pirozzi, G.; Graziano, A.; Galderisi, U.; Cavaliere, C.; De Rosa, A.; Papaccio, G.; et al. Detection and characterization of CD133+ cancer stem cells in human solid tumours. PLoS ONE 2008, 3, e3469. [Google Scholar] [CrossRef]
  27. Li, J.; Zhong, X.Y.; Li, Z.Y.; Cai, J.F.; Zou, L.; Li, J.M.; Yang, T.; Liu, W. CD133 expression in osteosarcoma and derivation of CD133(+) cells. Mol. Med. Rep. 2013, 7, 577–584. [Google Scholar] [CrossRef] [Green Version]
  28. He, A.; Qi, W.; Huang, Y.; Feng, T.; Chen, J.; Sun, Y.; Shen, Z.; Yao, Y. CD133 expression predicts lung metastasis and poor prognosis in osteosarcoma patients: A clinical and experimental study. Exp. Ther. Med. 2012, 4, 435–441. [Google Scholar] [CrossRef] [Green Version]
  29. Tirino, V.; Desiderio, V.; Paino, F.; De Rosa, A.; Papaccio, F.; Fazioli, F.; Pirozzi, G.; Papaccio, G. Human primary bone sarcomas contain CD133+ cancer stem cells displaying high tumorigenicity in vivo. FASEB J. 2011, 25, 2022–2030. [Google Scholar] [CrossRef]
  30. Fujiwara, T.; Katsuda, T.; Hagiwara, K.; Kosaka, N.; Yoshioka, Y.; Takahashi, R.U.; Takeshita, F.; Kubota, D.; Kondo, T.; Ichikawa, H.; et al. Clinical relevance and therapeutic significance of microRNA-133a expression profiles and functions in malignant osteosarcoma-initiating cells. Stem Cells 2014, 32, 959–973. [Google Scholar] [CrossRef]
  31. Adhikari, A.S.; Agarwal, N.; Wood, B.M.; Porretta, C.; Ruiz, B.; Pochampally, R.R.; Iwakuma, T. CD117 and Stro-1 identify osteosarcoma tumor-initiating cells associated with metastasis and drug resistance. Cancer Res. 2010, 70, 4602–4612. [Google Scholar] [CrossRef] [Green Version]
  32. Tian, J.; Li, X.; Si, M.; Liu, T.; Li, J. CD271+ osteosarcoma cells display stem-like properties. PLoS ONE 2014, 9, e98549. [Google Scholar] [CrossRef] [PubMed]
  33. Honoki, K.; Fujii, H.; Kubo, A.; Kido, A.; Mori, T.; Tanaka, Y.; Tsujiuchi, T. Possible involvement of stem-like populations with elevated ALDH1 in sarcomas for chemotherapeutic drug resistance. Oncol. Rep. 2010, 24, 501–505. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, L.; Park, P.; Zhang, H.; La Marca, F.; Lin, C.Y. Prospective identification of tumorigenic osteosarcoma cancer stem cells in OS99-1 cells based on high aldehyde dehydrogenase activity. Int. J. Cancer 2011, 128, 294–303. [Google Scholar] [CrossRef] [PubMed]
  35. Berman, S.D.; Calo, E.; Landman, A.S.; Danielian, P.S.; Miller, E.S.; West, J.C.; Fonhoue, B.D.; Caron, A.; Bronson, R.; Bouxsein, M.L.; et al. Metastatic osteosarcoma induced by inactivation of Rb and p53 in the osteoblast lineage. Proc. Natl. Acad. Sci. USA 2008, 105, 11851–11856. [Google Scholar] [CrossRef] [Green Version]
  36. Walkley, C.R.; Qudsi, R.; Sankaran, V.G.; Perry, J.A.; Gostissa, M.; Roth, S.I.; Rodda, S.J.; Snay, E.; Dunning, P.; Fahey, F.H.; et al. Conditional mouse osteosarcoma, dependent on p53 loss and potentiated by loss of Rb, mimics the human disease. Genes Dev. 2008, 22, 1662–1676. [Google Scholar] [CrossRef] [Green Version]
  37. Pan, Y.; Zhang, Y.; Tang, W.; Zhang, Y. Interstitial serum albumin empowers osteosarcoma cells with FAIM2 transcription to obtain viability via dedifferentiation. In Vitro Cell Dev. Biol. Anim. 2020, 56, 129–144. [Google Scholar] [CrossRef]
  38. Murase, M.; Kano, M.; Tsukahara, T.; Takahashi, A.; Torigoe, T.; Kawaguchi, S.; Kimura, S.; Wada, T.; Uchihashi, Y.; Kondo, T.; et al. Side population cells have the characteristics of cancer stem-like cells/cancer-initiating cells in bone sarcomas. Br. J. Cancer 2009, 101, 1425–1432. [Google Scholar] [CrossRef] [Green Version]
  39. Yang, M.; Yan, M.; Zhang, R.; Li, J.; Luo, Z. Side population cells isolated from human osteosarcoma are enriched with tumor-initiating cells. Cancer Sci. 2011, 102, 1774–1781. [Google Scholar] [CrossRef]
  40. Wang, Y.; Teng, J.S. Increased multi-drug resistance and reduced apoptosis in osteosarcoma side population cells are crucial factors for tumor recurrence. Exp. Ther. Med. 2016, 12, 81–86. [Google Scholar] [CrossRef]
  41. Rainusso, N.; Man, T.K.; Lau, C.C.; Hicks, J.; Shen, J.J.; Yu, A.; Wang, L.L.; Rosen, J.M. Identification and gene expression profiling of tumor-initiating cells isolated from human osteosarcoma cell lines in an orthotopic mouse model. Cancer Biol. 2011, 12, 278–287. [Google Scholar] [CrossRef]
  42. Martins-Neves, S.R.; Corver, W.E.; Paiva-Oliveira, D.I.; van den Akker, B.E.; Briaire-de-Bruijn, I.H.; Bovee, J.V.; Gomes, C.M.; Cleton-Jansen, A.M. Osteosarcoma Stem Cells Have Active Wnt/beta-catenin and Overexpress SOX2 and KLF4. J. Cell. Physiol. 2016, 231, 876–886. [Google Scholar] [CrossRef] [PubMed]
  43. Bonnet, D.; Dick, J.E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 1997, 3, 730–737. [Google Scholar] [CrossRef] [PubMed]
  44. Vinogradov, S.; Wei, X. Cancer stem cells and drug resistance: The potential of nanomedicine. Nanomedicine 2012, 7, 597–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Zhao, Y.; Alakhova, D.Y.; Kabanov, A.V. Can nanomedicines kill cancer stem cells? Adv. Drug Deliv. Rev. 2013, 65, 1763–1783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Jamieson, C.H.; Ailles, L.E.; Dylla, S.J.; Muijtjens, M.; Jones, C.; Zehnder, J.L.; Gotlib, J.; Li, K.; Manz, M.G.; Keating, A.; et al. Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML. N. Engl. J. Med. 2004, 351, 657–667. [Google Scholar] [CrossRef] [Green Version]
  47. Goardon, N.; Marchi, E.; Atzberger, A.; Quek, L.; Schuh, A.; Soneji, S.; Woll, P.; Mead, A.; Alford, K.A.; Rout, R.; et al. Coexistence of LMPP-like and GMP-like leukemia stem cells in acute myeloid leukemia. Cancer Cell 2011, 19, 138–152. [Google Scholar] [CrossRef] [Green Version]
  48. Gupta, P.B.; Fillmore, C.M.; Jiang, G.; Shapira, S.D.; Tao, K.; Kuperwasser, C.; Lander, E.S. Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells. Cell 2011, 146, 633–644. [Google Scholar] [CrossRef] [Green Version]
  49. Shackleton, M. Normal stem cells and cancer stem cells: Similar and different. Semin. Cancer Biol. 2010, 20, 85–92. [Google Scholar] [CrossRef]
  50. Sell, S. Stem cell origin of cancer and differentiation therapy. Crit. Rev. Oncol./Hematol. 2004, 51, 1–28. [Google Scholar] [CrossRef]
  51. Gener, P.; Seras-Franzoso, J.; Callejo, P.G.; Andrade, F.; Rafael, D.; Martínez, F.; Montero, S.; Arango, D.; Sayós, J.; Abasolo, I.; et al. Dynamism, Sensitivity, and Consequences of Mesenchymal and Stem-Like Phenotype of Cancer Cells. Stem Cells Int. 2018, 2018, 4516454. [Google Scholar] [CrossRef]
  52. Kreso, A.; Dick, J.E. Evolution of the cancer stem cell model. Cell Stem Cell 2014, 14, 275–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Shackleton, M.; Quintana, E.; Fearon, E.R.; Morrison, S.J. Heterogeneity in cancer: Cancer stem cells versus clonal evolution. Cell 2009, 138, 822–829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Prasad, P.; Mittal, S.A.; Chongtham, J.; Mohanty, S.; Srivastava, T. Hypoxia-Mediated Epigenetic Regulation of Stemness in Brain Tumor Cells. Stem Cells 2017, 35, 1468–1478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Gilchrist, K.W.; Gray, R.; Fowble, B.; Tormey, D.C.; Taylor, S.G.t. Tumor necrosis is a prognostic predictor for early recurrence and death in lymph node-positive breast cancer: A 10-year follow-up study of 728 Eastern Cooperative Oncology Group patients. J. Clin. Oncol. 1993, 11, 1929–1935. [Google Scholar] [CrossRef] [PubMed]
  56. Beck, B.; Driessens, G.; Goossens, S.; Youssef, K.K.; Kuchnio, A.; Caauwe, A.; Sotiropoulou, P.A.; Loges, S.; Lapouge, G.; Candi, A.; et al. A vascular niche and a VEGF-Nrp1 loop regulate the initiation and stemness of skin tumours. Nature 2011, 478, 399–403. [Google Scholar] [CrossRef] [PubMed]
  57. Zhang, B.; Li, Y.L.; Zhao, J.L.; Zhen, O.; Yu, C.; Yang, B.H.; Yu, X.R. Hypoxia-inducible factor-1 promotes cancer progression through activating AKT/Cyclin D1 signaling pathway in osteosarcoma. Biomed. Pharm. 2018, 105, 1–9. [Google Scholar] [CrossRef]
  58. Zhang, H.; Wu, H.; Zheng, J.; Yu, P.; Xu, L.; Jiang, P.; Gao, J.; Wang, H.; Zhang, Y. Transforming growth factor beta1 signal is crucial for dedifferentiation of cancer cells to cancer stem cells in osteosarcoma. Stem Cells 2013, 31, 433–446. [Google Scholar] [CrossRef]
  59. Maes, C.; Carmeliet, G.; Schipani, E. Hypoxia-driven pathways in bone development, regeneration and disease. Nat. Rev. Rheumatol. 2012, 8, 358–366. [Google Scholar] [CrossRef] [Green Version]
  60. Godet, I.; Shin, Y.J.; Ju, J.A.; Ye, I.C.; Wang, G.; Gilkes, D.M. Fate-mapping post-hypoxic tumor cells reveals a ROS-resistant phenotype that promotes metastasis. Nat. Commun. 2019, 10, 4862. [Google Scholar] [CrossRef]
  61. Maxwell, P.H.; Wiesener, M.S.; Chang, G.W.; Clifford, S.C.; Vaux, E.C.; Cockman, M.E.; Wykoff, C.C.; Pugh, C.W.; Maher, E.R.; Ratcliffe, P.J. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 1999, 399, 271–275. [Google Scholar] [CrossRef]
  62. Vanharanta, S.; Shu, W.; Brenet, F.; Hakimi, A.A.; Heguy, A.; Viale, A.; Reuter, V.E.; Hsieh, J.J.; Scandura, J.M.; Massagué, J. Epigenetic expansion of VHL-HIF signal output drives multiorgan metastasis in renal cancer. Nat. Med. 2013, 19, 50–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Mekhail, K.; Gunaratnam, L.; Bonicalzi, M.E.; Lee, S. HIF activation by pH-dependent nucleolar sequestration of VHL. Nat. Cell Biol. 2004, 6, 642–647. [Google Scholar] [CrossRef] [PubMed]
  64. Giaccia, A.J.; Simon, M.C.; Johnson, R. The biology of hypoxia: The role of oxygen sensing in development, normal function, and disease. Genes Dev. 2004, 18, 2183–2194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Rankin, E.B.; Giaccia, A.J. The role of hypoxia-inducible factors in tumorigenesis. Cell Death Differ. 2008, 15, 678–685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Lee, J.W.; Bae, S.H.; Jeong, J.W.; Kim, S.H.; Kim, K.W. Hypoxia-inducible factor (HIF-1)alpha: Its protein stability and biological functions. Exp. Mol. Med. 2004, 36, 1–12. [Google Scholar] [CrossRef] [Green Version]
  67. Ke, Q.; Costa, M. Hypoxia-inducible factor-1 (HIF-1). Mol. Pharmacol. 2006, 70, 1469–1480. [Google Scholar] [CrossRef]
  68. Koh, M.Y.; Powis, G. Passing the baton: The HIF switch. Trends Biochem. Sci. 2012, 37, 364–372. [Google Scholar] [CrossRef] [Green Version]
  69. Ouyang, Y.; Li, H.; Bu, J.; Li, X.; Chen, Z.; Xiao, T. Hypoxia-inducible factor-1 expression predicts osteosarcoma patients’ survival: A meta-analysis. Int. J. Biol. Mrk. 2016, 31, e229–e234. [Google Scholar] [CrossRef]
  70. Koka, P.; Mundre, R.S.; Rangarajan, R.; Chandramohan, Y.; Subramanian, R.K.; Dhanasekaran, A. Uncoupling Warburg effect and stemness in CD133(+ve) cancer stem cells from Saos-2 (osteosarcoma) cell line under hypoxia. Mol. Biol. Rep. 2018, 45, 1653–1662. [Google Scholar] [CrossRef]
  71. Lin, J.; Wang, X.; Wang, X.; Wang, S.; Shen, R.; Yang, Y.; Xu, J.; Lin, J. Hypoxia increases the expression of stem cell markers in human osteosarcoma cells. Oncol. Lett. 2021, 21, 217. [Google Scholar] [CrossRef]
  72. Zhang, H.; Mai, Q.; Chen, J. MicroRNA-210 is increased and it is required for dedifferentiation of osteosarcoma cell line. Cell Biol. Int. 2017, 41, 267–275. [Google Scholar] [CrossRef] [PubMed]
  73. Méndez, O.; Zavadil, J.; Esencay, M.; Lukyanov, Y.; Santovasi, D.; Wang, S.C.; Newcomb, E.W.; Zagzag, D. Knock down of HIF-1alpha in glioma cells reduces migration in vitro and invasion in vivo and impairs their ability to form tumor spheres. Mol. Cancer 2010, 9, 133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Couvelard, A.; O’Toole, D.; Turley, H.; Leek, R.; Sauvanet, A.; Degott, C.; Ruszniewski, P.; Belghiti, J.; Harris, A.L.; Gatter, K.; et al. Microvascular density and hypoxia-inducible factor pathway in pancreatic endocrine tumours: Negative correlation of microvascular density and VEGF expression with tumour progression. Br. J. Cancer 2005, 92, 94–101. [Google Scholar] [CrossRef]
  75. Pang, M.F.; Siedlik, M.J.; Han, S.; Stallings-Mann, M.; Radisky, D.C.; Nelson, C.M. Tissue Stiffness and Hypoxia Modulate the Integrin-Linked Kinase ILK to Control Breast Cancer Stem-like Cells. Cancer Res. 2016, 76, 5277–5287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Zhang, C.; Samanta, D.; Lu, H.; Bullen, J.W.; Zhang, H.; Chen, I.; He, X.; Semenza, G.L. Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m⁶A-demethylation of NANOG mRNA. Proc. Natl. Acad. Sci. USA 2016, 113, E2047–E2056. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Shi, Y.; Fan, S.; Wu, M.; Zuo, Z.; Li, X.; Jiang, L.; Shen, Q.; Xu, P.; Zeng, L.; Zhou, Y.; et al. YTHDF1 links hypoxia adaptation and non-small cell lung cancer progression. Nat. Commun. 2019, 10, 4892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Chae, Y.C.; Vaira, V.; Caino, M.C.; Tang, H.Y.; Seo, J.H.; Kossenkov, A.V.; Ottobrini, L.; Martelli, C.; Lucignani, G.; Bertolini, I.; et al. Mitochondrial Akt Regulation of Hypoxic Tumor Reprogramming. Cancer Cell 2016, 30, 257–272. [Google Scholar] [CrossRef] [Green Version]
  79. Gorgun, C.; Ozturk, S.; Gokalp, S.; Vatansever, S.; Gurhan, S.I.; Urkmez, A.S. Synergistic role of three dimensional niche and hypoxia on conservation of cancer stem cell phenotype. Int. J. Biol. Macromol. 2016, 90, 20–26. [Google Scholar] [CrossRef]
  80. Najafi, M.; Farhood, B.; Mortezaee, K.; Kharazinejad, E.; Majidpoor, J.; Ahadi, R. Hypoxia in solid tumors: A key promoter of cancer stem cell (CSC) resistance. J. Cancer Res. Clin. Oncol. 2020, 146, 19–31. [Google Scholar] [CrossRef] [PubMed]
  81. Sun, X.; Lv, X.; Yan, Y.; Zhao, Y.; Ma, R.; He, M.; Wei, M. Hypoxia-mediated cancer stem cell resistance and targeted therapy. Biomed. Pharm. 2020, 130, 110623. [Google Scholar] [CrossRef]
  82. Nathan, S.S.; DiResta, G.R.; Casas-Ganem, J.E.; Hoang, B.H.; Sowers, R.; Yang, R.; Huvos, A.G.; Gorlick, R.; Healey, J.H. Elevated physiologic tumor pressure promotes proliferation and chemosensitivity in human osteosarcoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2005, 11, 2389–2397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Pisanu, M.E.; Noto, A.; De Vitis, C.; Masiello, M.G.; Coluccia, P.; Proietti, S.; Giovagnoli, M.R.; Ricci, A.; Giarnieri, E.; Cucina, A.; et al. Lung cancer stem cell lose their stemness default state after exposure to microgravity. BioMed Res. Int. 2014, 2014, 470253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Lu, D.; Luo, C.; Zhang, C.; Li, Z.; Long, M. Differential regulation of morphology and stemness of mouse embryonic stem cells by substrate stiffness and topography. Biomaterials 2014, 35, 3945–3955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Hao, J.; Zhang, Y.; Ye, R.; Zheng, Y.; Zhao, Z.; Li, J. Mechanotransduction in cancer stem cells. Cell Biol. Int. 2013, 37, 888–891. [Google Scholar] [CrossRef]
  86. Tian, B.; Lin, W.; Zhang, Y. Effects of biomechanical forces on the biological behavior of cancer stem cells. J. Cancer 2021, 12, 5895–5902. [Google Scholar] [CrossRef]
  87. Northey, J.J.; Przybyla, L.; Weaver, V.M. Tissue Force Programs Cell Fate and Tumor Aggression. Cancer Discov. 2017, 7, 1224–1237. [Google Scholar] [CrossRef] [Green Version]
  88. Shieh, A.C. Biomechanical forces shape the tumor microenvironment. Ann. Biomed. Eng. 2011, 39, 1379–1389. [Google Scholar] [CrossRef]
  89. Mierke, C.T. The matrix environmental and cell mechanical properties regulate cell migration and contribute to the invasive phenotype of cancer cells. Rep. Prog. Phys. Phys. Soc. 2019, 82, 064602. [Google Scholar] [CrossRef]
  90. Plotkin, L.I.; Bellido, T. Osteocytic signalling pathways as therapeutic targets for bone fragility. Nat. Rev. Endocrinol. 2016, 12, 593–605. [Google Scholar] [CrossRef]
  91. You, Y.; Zheng, Q.; Dong, Y.; Xie, X.; Wang, Y.; Wu, S.; Zhang, L.; Wang, Y.; Xue, T.; Wang, Z.; et al. Matrix stiffness-mediated effects on stemness characteristics occurring in HCC cells. Oncotarget 2016, 7, 32221–32231. [Google Scholar] [CrossRef] [Green Version]
  92. Tan, F.; Huang, Y.; Pei, Q.; Liu, H.; Pei, H.; Zhu, H. Matrix stiffness mediates stemness characteristics via activating the Yes-associated protein in colorectal cancer cells. J. Cell. Biochem. 2018, 120, 2213–2225. [Google Scholar] [CrossRef] [PubMed]
  93. Tian, B.; Luo, Q.; Ju, Y.; Song, G. A Soft Matrix Enhances the Cancer Stem Cell Phenotype of HCC Cells. Int. J. Mol. Sci. 2019, 20, 2831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Tan, Y.; Tajik, A.; Chen, J.; Jia, Q.; Chowdhury, F.; Wang, L.; Chen, J.; Zhang, S.; Hong, Y.; Yi, H.; et al. Matrix softness regulates plasticity of tumour-repopulating cells via H3K9 demethylation and Sox2 expression. Nat. Commun. 2014, 5, 4619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Ip, C.K.; Li, S.S.; Tang, M.Y.; Sy, S.K.; Ren, Y.; Shum, H.C.; Wong, A.S. Stemness and chemoresistance in epithelial ovarian carcinoma cells under shear stress. Sci. Rep. 2016, 6, 26788. [Google Scholar] [CrossRef] [Green Version]
  96. Triantafillu, U.L.; Park, S.; Klaassen, N.L.; Raddatz, A.D.; Kim, Y. Fluid shear stress induces cancer stem cell-like phenotype in MCF7 breast cancer cell line without inducing epithelial to mesenchymal transition. Int. J. Oncol. 2017, 50, 993–1001. [Google Scholar] [CrossRef] [Green Version]
  97. Sun, J.; Luo, Q.; Liu, L.; Song, G. Low-level shear stress induces differentiation of liver cancer stem cells via the Wnt/beta-catenin signalling pathway. Exp. Cell Res. 2018, 397, 90–96. [Google Scholar] [CrossRef]
  98. Sun, J.; Luo, Q.; Liu, L.; Song, G. Low-level shear stress promotes migration of liver cancer stem cells via the FAK-ERK1/2 signalling pathway. Cancer Lett. 2018, 427, 1–8. [Google Scholar] [CrossRef]
  99. Li, S.; Bai, H.; Chen, X.; Gong, S.; Xiao, J.; Li, D.; Li, L.; Jiang, Y.; Li, T.; Qin, X.; et al. Soft Substrate Promotes Osteosarcoma Cell Self-Renewal, Differentiation, and Drug Resistance Through miR-29b and Its Target Protein Spin 1. ACS Biomater. Sci. Eng. 2020, 6, 5588–5598. [Google Scholar] [CrossRef]
  100. Mitra, A.; Mishra, L.; Li, S. EMT, CTCs and CSCs in tumor relapse and drug-resistance. Oncotarget 2015, 6, 10697–10711. [Google Scholar] [CrossRef]
  101. Mani, S.A.; Guo, W.; Liao, M.J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.; Shipitsin, M.; et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008, 133, 704–715. [Google Scholar] [CrossRef] [Green Version]
  102. 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] [PubMed]
  103. Gupta, R.K.; Johansson, S. beta1 integrins restrict the growth of foci and spheroids. Histochem. Cell Biol. 2012, 138, 881–894. [Google Scholar] [CrossRef] [PubMed]
  104. Schrader, J.; Gordon-Walker, T.T.; Aucott, R.L.; van Deemter, M.; Quaas, A.; Walsh, S.; Benten, D.; Forbes, S.J.; Wells, R.G.; Iredale, J.P. Matrix stiffness modulates proliferation, chemotherapeutic response, and dormancy in hepatocellular carcinoma cells. Hepatology 2011, 53, 1192–1205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Bu, Y.; Cao, D. The origin of cancer stem cells. Front. Biosci. 2012, 4, 819–830. [Google Scholar]
  106. Lowery, F.J.; Yu, D. Growth factor signaling in metastasis: Current understanding and future opportunities. Cancer Metastasis Rev. 2012, 31, 479–491. [Google Scholar] [CrossRef]
  107. Lopez de Andres, J.; Grinan-Lison, C.; Jimenez, G.; Marchal, J.A. Cancer stem cell secretome in the tumor microenvironment: A key point for an effective personalized cancer treatment. J. Hematol. Oncol. 2020, 13, 136. [Google Scholar] [CrossRef]
  108. Lamora, A.; Talbot, J.; Bougras, G.; Amiaud, J.; Leduc, M.; Chesneau, J.; Taurelle, J.; Stresing, V.; Le Deley, M.C.; Heymann, M.F.; et al. Overexpression of smad7 blocks primary tumor growth and lung metastasis development in osteosarcoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2014, 20, 5097–5112. [Google Scholar] [CrossRef] [Green Version]
  109. Peng, D.; Fu, M.; Wang, M.; Wei, Y.; Wei, X. Targeting TGF-beta signal transduction for fibrosis and cancer therapy. Mol. Cancer 2022, 21, 104. [Google Scholar] [CrossRef]
  110. Wang, T.; Wang, D.; Zhang, L.; Yang, P.; Wang, J.; Liu, Q.; Yan, F.; Lin, F. The TGFbeta-miR-499a-SHKBP1 pathway induces resistance to EGFR inhibitors in osteosarcoma cancer stem cell-like cells. J. Exp. Clin. Cancer Res. CR 2019, 38, 226. [Google Scholar] [CrossRef] [Green Version]
  111. Matsumoto, T.; Yokoi, A.; Hashimura, M.; Oguri, Y.; Akiya, M.; Saegusa, M. TGF-β-mediated LEFTY/Akt/GSK-3β/Snail axis modulates epithelial-mesenchymal transition and cancer stem cell properties in ovarian clear cell carcinomas. Mol. Carcinog. 2018, 57, 957–967. [Google Scholar] [CrossRef]
  112. Zhang, B.; Ye, H.; Ren, X.; Zheng, S.; Zhou, Q.; Chen, C.; Lin, Q.; Li, G.; Wei, L.; Fu, Z.; et al. Macrophage-expressed CD51 promotes cancer stem cell properties via the TGF-β1/smad2/3 axis in pancreatic cancer. Cancer Lett. 2019, 459, 204–215. [Google Scholar] [CrossRef] [PubMed]
  113. Wang, L.; Park, P.; Zhang, H.; La Marca, F.; Claeson, A.; Valdivia, J.; Lin, C.-Y. BMP-2 inhibits the tumorigenicity of cancer stem cells in human osteosarcoma OS99-1 cell line. Cancer Biol. Ther. 2014, 11, 457–463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Zhang, W.; Zhao, J.M.; Lin, J.; Hu, C.Z.; Zhang, W.B.; Yang, W.L.; Zhang, J.; Zhang, J.W.; Zhu, J. Adaptive Fibrogenic Reprogramming of Osteosarcoma Stem Cells Promotes Metastatic Growth. Cell Rep. 2018, 24, 1266–1277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Yao, J.; Lin, J.; He, L.; Huang, J.; Liu, Q. TNF-alpha/miR-155 axis induces the transformation of osteosarcoma cancer stem cells independent of TP53INP1. Gene 2020, 726, 144224. [Google Scholar] [CrossRef] [PubMed]
  116. 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]
  117. Mao, X.; Xu, J.; Wang, W.; Liang, C.; Hua, J.; Liu, J.; Zhang, B.; Meng, Q.; Yu, X.; Shi, S. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: New findings and future perspectives. Mol. Cancer 2021, 20, 131. [Google Scholar] [CrossRef]
  118. Melzer, C.; von der Ohe, J.; Lehnert, H.; Ungefroren, H.; Hass, R. Cancer stem cell niche models and contribution by mesenchymal stroma/stem cells. Mol. Cancer 2017, 16, 28. [Google Scholar] [CrossRef] [Green Version]
  119. Cortini, M.; Massa, A.; Avnet, S.; Bonuccelli, G.; Baldini, N. Tumor-Activated Mesenchymal Stromal Cells Promote Osteosarcoma Stemness and Migratory Potential via IL-6 Secretion. PLoS ONE 2016, 11, e0166500. [Google Scholar] [CrossRef] [Green Version]
  120. Zhao, Y.; Zhang, B.; Zhang, Q.; Ma, X.; Feng, H. Tumor-associated macrophages in osteosarcoma. J. Zhejiang Univ. Sci. B 2021, 22, 885–892. [Google Scholar] [CrossRef]
  121. Shao, X.J.; Xiang, S.F.; Chen, Y.Q.; Zhang, N.; Cao, J.; Zhu, H.; Yang, B.; Zhou, Q.; Ying, M.D.; He, Q.J. Inhibition of M2-like macrophages by all-trans retinoic acid prevents cancer initiation and stemness in osteosarcoma cells. Acta Pharmacol. Sin. 2019, 40, 1343–1350. [Google Scholar] [CrossRef]
  122. Lu, H.; Clauser, K.R.; Tam, W.L.; Fröse, J.; Ye, X.; Eaton, E.N.; Reinhardt, F.; Donnenberg, V.S.; Bhargava, R.; Carr, S.A.; et al. A breast cancer stem cell niche supported by juxtacrine signalling from monocytes and macrophages. Nat. Cell Biol. 2014, 16, 1105–1117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Korkaya, H.; Liu, S.; Wicha, M.S. Regulation of cancer stem cells by cytokine networks: Attacking cancer’s inflammatory roots. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2011, 17, 6125–6129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. O’Brien, K.; Breyne, K.; Ughetto, S.; Laurent, L.C.; Breakefield, X.O. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat. Rev. Mol. Cell Biol. 2020, 21, 585–606. [Google Scholar] [CrossRef]
  125. Skotland, T.; Sagini, K.; Sandvig, K.; Llorente, A. An emerging focus on lipids in extracellular vesicles. Adv. Drug Deliv. Rev. 2020, 159, 308–321. [Google Scholar] [CrossRef] [PubMed]
  126. Yang, Q.; Liu, J.; Wu, B.; Wang, X.; Jiang, Y.; Zhu, D. Role of extracellular vesicles in osteosarcoma. Int. J. Med. Sci. 2022, 19, 1216–1226. [Google Scholar] [CrossRef]
  127. De Martino, V.; Rossi, M.; Battafarano, G.; Pepe, J.; Minisola, S.; Del Fattore, A. Extracellular Vesicles in Osteosarcoma: Antagonists or Therapeutic Agents? Int. J. Mol. Sci. 2021, 22, 12586. [Google Scholar] [CrossRef]
  128. Sarhadi, V.K.; Daddali, R.; Seppanen-Kaijansinkko, R. Mesenchymal Stem Cells and Extracellular Vesicles in Osteosarcoma Pathogenesis and Therapy. Int. J. Mol. Sci. 2021, 22, 11035. [Google Scholar] [CrossRef]
  129. Li, S. The basic characteristics of extracellular vesicles and their potential application in bone sarcomas. J. Nanobiotechnol. 2021, 19, 277. [Google Scholar] [CrossRef]
  130. Yang, J.; Hu, Y.; Wang, L.; Sun, X.; Yu, L.; Guo, W. Human umbilical vein endothelial cells derived-exosomes promote osteosarcoma cell stemness by activating Notch signaling pathway. Bioengineered 2021, 12, 11007–11017. [Google Scholar] [CrossRef]
  131. Zhang, H.; Wang, J.; Ren, T.; Huang, Y.; Liang, X.; Yu, Y.; Wang, W.; Niu, J.; Guo, W. Bone marrow mesenchymal stem cell-derived exosomal miR-206 inhibits osteosarcoma progression by targeting TRA2B. Cancer Lett. 2020, 490, 54–65. [Google Scholar] [CrossRef]
  132. He, H.; Ding, M.; Li, T.; Zhao, W.; Zhang, L.; Yin, P.; Zhang, W. Bone mesenchymal stem cell-derived extracellular vesicles containing NORAD promote osteosarcoma by miR-30c-5p. Lab. Investig. 2022, 102, 826–837. [Google Scholar] [CrossRef] [PubMed]
  133. Chang, J.; Li, H.; Zhu, Z.; Mei, P.; Hu, W.; Xiong, X.; Tao, J. microRNA-21-5p from M2 macrophage-derived extracellular vesicles promotes the differentiation and activity of pancreatic cancer stem cells by mediating KLF3. Cell Biol. Toxicol. 2022, 38, 577–590. [Google Scholar] [CrossRef] [PubMed]
  134. Chung, W.M.; Molony, R.D.; Lee, Y.F. Non-stem bladder cancer cell-derived extracellular vesicles promote cancer stem cell survival in response to chemotherapy. Stem Cell Res. Ther. 2021, 12, 533. [Google Scholar] [CrossRef]
  135. Brossa, A.; Fonsato, V.; Grange, C.; Tritta, S.; Tapparo, M.; Calvetti, R.; Cedrino, M.; Fallo, S.; Gontero, P.; Camussi, G.; et al. Extracellular vesicles from human liver stem cells inhibit renal cancer stem cell-derived tumor growth in vitro and in vivo. Int. J. Cancer 2020, 147, 1694–1706. [Google Scholar] [CrossRef] [Green Version]
  136. Zhang, H.G.; Grizzle, W.E. Exosomes: A novel pathway of local and distant intercellular communication that facilitates the growth and metastasis of neoplastic lesions. Am. J. Pathol. 2014, 184, 28–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Zhao, H.; Chen, S.; Fu, Q. Exosomes from CD133(+) cells carrying circ-ABCC1 mediate cell stemness and metastasis in colorectal cancer. J. Cell. Biochem. 2020, 121, 3286–3297. [Google Scholar] [CrossRef]
  138. Li, W.; Zhang, L.; Guo, B.; Deng, J.; Wu, S.; Li, F.; Wang, Y.; Lu, J.; Zhou, Y. Exosomal FMR1-AS1 facilitates maintaining cancer stem-like cell dynamic equilibrium via TLR7/NFκB/c-Myc signaling in female esophageal carcinoma. Mol. Cancer 2019, 18, 22. [Google Scholar] [CrossRef] [Green Version]
  139. Sun, Z.; Wang, L.; Zhou, Y.; Dong, L.; Ma, W.; Lv, L.; Zhang, J.; Wang, X. Glioblastoma Stem Cell-Derived Exosomes Enhance Stemness and Tumorigenicity of Glioma Cells by Transferring Notch1 Protein. Cell. Mol. Neurobiol. 2020, 40, 767–784. [Google Scholar] [CrossRef]
  140. 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. Pharm. 2020, 127, 110141. [Google Scholar] [CrossRef]
  141. Mercer, T.R.; Munro, T.; Mattick, J.S. The potential of long noncoding RNA therapies. Trends Pharmacol. Sci. 2022, 43, 269–280. [Google Scholar] [CrossRef]
  142. Gebert, L.F.R.; MacRae, I.J. Regulation of microRNA function in animals. Nat. Rev. Mol. Cell Biol. 2019, 20, 21–37. [Google Scholar] [CrossRef] [PubMed]
  143. Ghafouri-Fard, S.; Shirvani-Farsani, Z.; Hussen, B.M.; Taheri, M. The critical roles of lncRNAs in the development of osteosarcoma. Biomed. Pharm. 2021, 135, 111217. [Google Scholar] [CrossRef] [PubMed]
  144. Liu, J.; Shang, G. The Roles of Noncoding RNAs in the Development of Osteosarcoma Stem Cells and Potential Therapeutic Targets. Front. Cell Dev. Biol. 2022, 10, 773038. [Google Scholar] [CrossRef] [PubMed]
  145. Zhang, Y.; Pan, Y.; Xie, C.; Zhang, Y. miR-34a exerts as a key regulator in the dedifferentiation of osteosarcoma via PAI-1-Sox2 axis. Cell Death Dis. 2018, 9, 777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Lu, J.; Song, G.; Tang, Q.; Yin, J.; Zou, C.; Zhao, Z.; Xie, X.; Xu, H.; Huang, G.; Wang, J.; et al. MiR-26a inhibits stem cell-like phenotype and tumor growth of osteosarcoma by targeting Jagged1. Oncogene 2017, 36, 231–241. [Google Scholar] [CrossRef] [PubMed]
  147. Zou, Y.; Huang, Y.; Yang, J.; Wu, J.; Luo, C. miR-34a is downregulated in human osteosarcoma stem-like cells and promotes invasion, tumorigenic ability and self-renewal capacity. Mol. Med. Rep. 2017, 15, 1631–1637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Liang, X.; Xu, C.; Wang, W.; Li, X. The DNMT1/miR-34a Axis Is Involved in the Stemness of Human Osteosarcoma Cells and Derived Stem-Like Cells. Stem Cells Int. 2019, 2019, 7028901. [Google Scholar] [CrossRef]
  149. Zhao, F.; Lv, J.; Gan, H.; Li, Y.; Wang, R.; Zhang, H.; Wu, Q.; Chen, Y. MiRNA profile of osteosarcoma with CD117 and stro-1 expression: miR-1247 functions as an onco-miRNA by targeting MAP3K9. Int. J. Clin. Exp. Pathol. 2015, 8, 1451–1458. [Google Scholar]
  150. 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. J. Am. Soc. Gene Ther. 2015, 23, 89–98. [Google Scholar] [CrossRef] [Green Version]
  151. Di Fiore, R.; Drago-Ferrante, R.; Pentimalli, F.; Di Marzo, D.; Forte, I.M.; D’Anneo, A.; Carlisi, D.; De Blasio, A.; Giuliano, M.; Tesoriere, G.; et al. MicroRNA-29b-1 impairs in vitro cell proliferation, self-renewal and chemoresistance of human osteosarcoma 3AB-OS cancer stem cells. Int. J. Oncol. 2014, 45, 2013–2023. [Google Scholar] [CrossRef] [Green Version]
  152. Guo, X.; Yu, L.; Zhang, Z.; Dai, G.; Gao, T.; Guo, W. miR-335 negatively regulates osteosarcoma stem cell-like properties by targeting POU5F1. Cancer Cell Int. 2017, 17, 29. [Google Scholar] [CrossRef] [PubMed]
  153. Jiang, N.; Wang, X.; Xie, X.; Liao, Y.; Liu, N.; Liu, J.; Miao, N.; Shen, J.; Peng, T. lncRNA DANCR promotes tumor progression and cancer stemness features in osteosarcoma by upregulating AXL via miR-33a-5p inhibition. Cancer Lett. 2017, 405, 46–55. [Google Scholar] [CrossRef] [PubMed]
  154. Zhang, R.M.; Tang, T.; Yu, H.M.; Yao, X.D. LncRNA DLX6-AS1/miR-129-5p/DLK1 axis aggravates stemness of osteosarcoma through Wnt signaling. Biochem. Biophys. Res. Commun. 2018, 507, 260–266. [Google Scholar] [CrossRef] [PubMed]
  155. Wang, Y.; Yao, J.; Meng, H.; Yu, Z.; Wang, Z.; Yuan, X.; Chen, H.; Wang, A. A novel long non-coding RNA, hypoxia-inducible factor-2alpha promoter upstream transcript, functions as an inhibitor of osteosarcoma stem cells in vitro. Mol. Med. Rep. 2015, 11, 2534–2540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Wu, H.; He, Y.; Chen, H.; Liu, Y.; Wei, B.; Chen, G.; Lin, H.; Lin, H. LncRNA THOR increases osteosarcoma cell stemness and migration by enhancing SOX9 mRNA stability. FEBS Open Bio 2019, 9, 781–790. [Google Scholar] [CrossRef] [Green Version]
  157. 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]
  158. Kong, Y.; Nie, Z.; Guo, H.; Ma, C. LINK-A lncRNA is upregulated in osteosarcoma and regulates migration, invasion and stemness of osteosarcoma cells. Oncol. Lett. 2020, 19, 2832–2838. [Google Scholar] [CrossRef]
  159. Chen, Y.; Huang, W.; Sun, W.; Zheng, B.; Wang, C.; Luo, Z.; Wang, J.; Yan, W. LncRNA MALAT1 Promotes Cancer Metastasis in Osteosarcoma via Activation of the PI3K-Akt Signaling Pathway. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2018, 51, 1313–1326. [Google Scholar] [CrossRef]
  160. Wang, Z.; Tan, M.; Chen, G.; Li, Z.; Lu, X. LncRNA SOX2-OT is a novel prognostic biomarker for osteosarcoma patients and regulates osteosarcoma cells proliferation and motility through modulating SOX2. IUBMB Life 2017, 69, 867–876. [Google Scholar] [CrossRef] [Green Version]
  161. Li, Z.; Wang, Y.; Hu, R.; Xu, R.; Xu, W. LncRNA B4GALT1-AS1 recruits HuR to promote osteosarcoma cells stemness and migration via enhancing YAP transcriptional activity. Cell Prolif. 2018, 51, e12504. [Google Scholar] [CrossRef] [Green Version]
  162. Shi, P.; Li, Y.; Guo, Q. Circular RNA circPIP5K1A contributes to cancer stemness of osteosarcoma by miR-515-5p/YAP axis. J. Transl. Med. 2021, 19, 464. [Google Scholar] [CrossRef] [PubMed]
  163. Kammertoens, T.; Schüler, T.; Blankenstein, T. Immunotherapy: Target the stroma to hit the tumor. Trends Mol. Med. 2005, 11, 225–231. [Google Scholar] [CrossRef] [PubMed]
  164. Fulda, S. Regulation of apoptosis pathways in cancer stem cells. Cancer Lett. 2013, 338, 168–173. [Google Scholar] [CrossRef] [PubMed]
  165. Martins-Neves, S.R.; Paiva-Oliveira, D.I.; Wijers-Koster, P.M.; Abrunhosa, A.J.; Fontes-Ribeiro, C.; Bovée, J.V.; Cleton-Jansen, A.M.; Gomes, C.M. Chemotherapy induces stemness in osteosarcoma cells through activation of Wnt/β-catenin signaling. Cancer Lett. 2016, 370, 286–295. [Google Scholar] [CrossRef] [PubMed]
  166. Xu, Z.; Wang, Y.; Zhang, L.; Huang, L. Nanoparticle-delivered transforming growth factor-β siRNA enhances vaccination against advanced melanoma by modifying tumor microenvironment. ACS Nano 2014, 8, 3636–3645. [Google Scholar] [CrossRef]
  167. Colak, S.; Ten Dijke, P. Targeting TGF-beta Signaling in Cancer. Trends Cancer 2017, 3, 56–71. [Google Scholar] [CrossRef]
  168. Piccirillo, S.G.; Reynolds, B.A.; Zanetti, N.; Lamorte, G.; Binda, E.; Broggi, G.; Brem, H.; Olivi, A.; Dimeco, F.; Vescovi, A.L. Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells. Nature 2006, 444, 761–765. [Google Scholar] [CrossRef]
  169. Chen, Y.; Cao, J.; Zhang, N.; Yang, B.; He, Q.; Shao, X.; Ying, M. Advances in differentiation therapy for osteosarcoma. Drug Discov. Today 2020, 25, 497–504. [Google Scholar] [CrossRef]
  170. Blackhall, F.; Jao, K.; Greillier, L.; Cho, B.C.; Penkov, K.; Reguart, N.; Majem, M.; Nackaerts, K.; Syrigos, K.; Hansen, K.; et al. Efficacy and Safety of Rovalpituzumab Tesirine Compared With Topotecan as Second-Line Therapy in DLL3-High SCLC: Results From the Phase 3 TAHOE Study. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2021, 16, 1547–1558. [Google Scholar] [CrossRef]
  171. Tang, Q.L.; Zhao, Z.Q.; Li, J.C.; Liang, Y.; Yin, J.Q.; Zou, C.Y.; Xie, X.B.; Zeng, Y.X.; Shen, J.N.; Kang, T.; et al. Salinomycin inhibits osteosarcoma by targeting its tumor stem cells. Cancer Lett. 2011, 311, 113–121. [Google Scholar] [CrossRef]
  172. Bayik, D.; Lathia, J.D. Cancer stem cell-immune cell crosstalk in tumour progression. Nat. Rev. Cancer 2021, 21, 526–536. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Role of microenvironment signaling in regulating OSC and non-OSC reversion. Complex pathways are necessary for the maintenance of the homeostasis of the OSC population. All microenvironment components, including cells (mesenchymal cells and immune cells) and non-cellular factors (hypoxia, cytokines and mechanical EVs), can influence the dynamic transition between OSCs and non-OSCs. OSCs, osteosarcoma stem cells; miR, microRNA; HIF, hypoxia-inducible factor; EVs, extracellular vesicles; EMT, epithelial-mesenchymal transition; BMP, bone morphogenetic protein; TNF, tumor necrosis factor; TGF, transforming growth factor; TRA2B, transformer 2β homolog.
Figure 1. Role of microenvironment signaling in regulating OSC and non-OSC reversion. Complex pathways are necessary for the maintenance of the homeostasis of the OSC population. All microenvironment components, including cells (mesenchymal cells and immune cells) and non-cellular factors (hypoxia, cytokines and mechanical EVs), can influence the dynamic transition between OSCs and non-OSCs. OSCs, osteosarcoma stem cells; miR, microRNA; HIF, hypoxia-inducible factor; EVs, extracellular vesicles; EMT, epithelial-mesenchymal transition; BMP, bone morphogenetic protein; TNF, tumor necrosis factor; TGF, transforming growth factor; TRA2B, transformer 2β homolog.
Ijms 23 16155 g001
Figure 2. Key non-coding RNAs in non-OSC and OSC interconversion signaling. A series of studies have reported that a number of non-coding RNAs, including microRNAs, lncRNAs and circRNAs, can participate in the OSC interconversion mechanism. OSC, osteosarcoma stem cell; lncRNA, long non-coding RNA; circRNA, circular RNA; AS1, antisense 1; B4GALT1, β-1,4-galactosyltransferase 1; DANCR, differentiation antagonizing non-protein coding RNA; LINK-A, long intergenic non-coding RNA for kinase activation; MALAT1, metastasis-associated lung adenocarcinoma transcript 1; Sex-determining region Y-box 2 overlapping transcript; THOR, testis-associated oncogenic lncRNA; NIRP1, Nuclear receptor interacting protein 1; PIP5K1A, Phosphatidylinositol-4-Phosphate 5-Kinase Type 1α; let, lethal; FER1L4, fer-1 family member 4; HIF2PUT, hypoxia-inducible factor-2α promoter upstream transcript.
Figure 2. Key non-coding RNAs in non-OSC and OSC interconversion signaling. A series of studies have reported that a number of non-coding RNAs, including microRNAs, lncRNAs and circRNAs, can participate in the OSC interconversion mechanism. OSC, osteosarcoma stem cell; lncRNA, long non-coding RNA; circRNA, circular RNA; AS1, antisense 1; B4GALT1, β-1,4-galactosyltransferase 1; DANCR, differentiation antagonizing non-protein coding RNA; LINK-A, long intergenic non-coding RNA for kinase activation; MALAT1, metastasis-associated lung adenocarcinoma transcript 1; Sex-determining region Y-box 2 overlapping transcript; THOR, testis-associated oncogenic lncRNA; NIRP1, Nuclear receptor interacting protein 1; PIP5K1A, Phosphatidylinositol-4-Phosphate 5-Kinase Type 1α; let, lethal; FER1L4, fer-1 family member 4; HIF2PUT, hypoxia-inducible factor-2α promoter upstream transcript.
Ijms 23 16155 g002
Table 1. Putative OSC markers and phenotypes.
Table 1. Putative OSC markers and phenotypes.
MarkerCell OriginPhenotype
CD133Saos-2, MG-63, U2-OS, MNNG/HOS, 143B, HOS, Human primary cellsHigh stem cells gene expression, sphere formation, side population, increased cell proliferation [26,27,28,29,30].
CD117/Stro-1K7M2, KHOS/NP, MNNG/HOS, 318–1, P932, BCOSHigh stem cells gene expression, sphere formation, drug resistance, in vivo tumorigenicity and metastatic potential [17,18,19,20,21,22,23,24,25,26,27,28,29,30,31].
CD271Human primary (FFPE), MNNG/HOS, U2-OS, Saos-2High stem cells gene expression, sphere formation, drug resistance, in vivo tumorigenicity [32].
Aldehyde dehydrogenaseMG-63, OS99–1 Hu09, Saos-2High stem cells gene expression, sphere formation, drug resistance, increased cell proliferation [33,34].
Stem cells antigen-14 Murine osteosarcoma cell linesSphere formation, in vivo tumorigenicity [35,36]
Fas apoptotic inhibitory molecule 2MNNG/HOS, U2-OSSphere formation, drug resistance, in vivo tumorigenicity [37].
Side populationOS2000, KIKU, NY, Huo9,
HOS, U2OS, Saos-2, human primary
High stem cells gene expression, Sphere formation, in vivo tumorigenicity, self-renewal, apoptosis resistant [38,39,40].
Sphere formationMG-63, MNNG/HOS, human primaryHigh stem cells gene expression, drug resistance, in vivo tumorigenicity [17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,41,42].
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tian, B.; Du, X.; Zheng, S.; Zhang, Y. The Role of Tumor Microenvironment in Regulating the Plasticity of Osteosarcoma Cells. Int. J. Mol. Sci. 2022, 23, 16155. https://doi.org/10.3390/ijms232416155

AMA Style

Tian B, Du X, Zheng S, Zhang Y. The Role of Tumor Microenvironment in Regulating the Plasticity of Osteosarcoma Cells. International Journal of Molecular Sciences. 2022; 23(24):16155. https://doi.org/10.3390/ijms232416155

Chicago/Turabian Style

Tian, Boren, Xiaoyun Du, Shiyu Zheng, and Yan Zhang. 2022. "The Role of Tumor Microenvironment in Regulating the Plasticity of Osteosarcoma Cells" International Journal of Molecular Sciences 23, no. 24: 16155. https://doi.org/10.3390/ijms232416155

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