Role of exosomal miR‑21 in the tumor microenvironment and osteosarcoma tumorigenesis and progression (Review)

  • Authors:
    • Shoufeng Wang
    • Fang Ma
    • Yi Feng
    • Tang Liu
    • Shasha He
  • View Affiliations

  • Published online on: February 19, 2020     https://doi.org/10.3892/ijo.2020.4992
  • Pages: 1055-1063
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Osteosarcoma is the most common bone tumor affecting both adolescents and children. Early detection is critical for the effective treatment of the disease. Derived from cancer cells, miR‑21 contained within exosomes in the tumor microenvironment may act on both cancer cells and the surrounding tumor microenvironment (TME), including immune cells, endothelial cells and fibroblasts. In human serum and plasm, the level of exosomal miR‑21 between osteosarcoma patients and healthy controls differs, supporting the role of miR‑21 as a biomarker for osteosarcoma. The involvement of a number of miR‑21 target genes in tumor progression suggests that miR‑21 may significantly affect the plasticity of cancer cells, leading to tumor progression, metastasis, angiogenesis and immune escape in osteosarcoma. Understanding the biogenesis and functions of exosomal miR‑21 is of great value for the diagnosis and therapy of cancer, including osteosarcoma. The present review discusses the role of miR‑21 in the tumor microenvironment, and in the development and progression of osteosarcoma, with an aim to summarize the functions of this miRNA in cancer.

1. Osteosarcoma

Osteosarcoma is the most common human primary malignant bone tumor affecting children and young adults (1), and is usually located in the distal femur, the proximal tibia, or the proximal humerus (2). The tumor is evolved from mesenchymal cells and is characterized by spindle cells and aberrant osteoid formation pathologically (3). The survival of patients with localized disease is increased with aggressive, multi-agent, neo-adjuvant chemotherapy and limb-salvage surgery. However, despite aggressive therapies, the long-term survival rate of patients with metastatic or recurrent disease however is <20% (2,4,5). Although several genes have been reported to be involved in osteosarcoma tumorigenesis and therapeutic resistance (6-11), the precise molecular mechanisms involved in these processes remain unclear. The identification of clinically relevant diagnostic and prognostic biomarkers, such as circulating or cellular/tissue biomarkers, is, therefore, urgently required.

2. Exosomes

Exosomes are lipid bilayer membrane bound, nano-sized extracellular vesicles that are 40-100 nm in diameter (12-14). The exosomes are formed by pinching off of multivesicular endosomes (MVEs) on the membrane in the form of small intraluminal vesicles within the MVE and packing with cytoplasmic contents, including proteins, mRNAs and microRNAs (miRNAs or miRs) (15,16). The secretion of exosomes occurs when MVEs fuse with the plasma membrane, and thus release the contents in exosomes into the extracellular environment. According to the exosome content database, ExoCarta, 9,769 proteins, 1,116 lipids, 3,408 mRNAs and 2,838 miRNAs have been identified in exosomes of different cell types in multiple organisms (17,18).

Exosomes were first described as vesicles released from reticulocyte MVEs for the removal of obsolete transferrin receptors (19,20). However, in recent years, exosomes have been considered to be important mediators of cellular communication in both normal physiological processes, and in the development and progression of diseases, such as cancer. Exosomes have been identified in the majority of bodily fluids, including the serum, urine, amniotic fluid, saliva, breastmilk, cerebrospinal fluid and nasal secretions (21,22). Importantly, cancer cells secrete a greater number of exosomes compared to healthy cells (23), indicating their potential for use as diagnostic biomarkers.

3. Exosomal miRNAs and cancer

miRNAs are a class of small non-coding endogenous RNAs (18-24 nucleotides in length). miRNAs can act as post-transcriptional gene regulators by pairing with complementary sequences in the 3′ untranslated region (3′ UTR) of target mRNAs, leading to mRNA degradation or translational repression (24,25). miRNAs regulate a variety of cellular processes associated with carcinogenesis, such as cell proliferation, cell cycle, apoptosis, angiogenesis, invasion and metastasis (26-31).

The expression of miRNAs is altered in a variety of cancer types and is associated with the disease stage in some cases. Some specific miRNAs may contribute to tumor growth, progression, metastasis and drug resistance (32-35). Among these, miR-21 is most notable, since it has been extensively studied in various types of cancer. The majority of miRNAs detectable in serum and saliva is concentrated within exosomes (23). The level of miRNAs is similar in both circulating exosomes from cancer patients and tumor cells (22,36). In a number of studies, the high expression of circulating miR-21 has been used to differentiate cancer patients from healthy individuals and predict disease outcomes (37 and refs. therein), suggesting that circulating exosome miRNAs, such as miR-21 can be utilized as for liquid biopsy miRNA profiling.

4. Exosomal miR-21 and osteosarcoma

hsa-mir-21 or miR-21 is an abundantly expressed miRNA in different types of mammalian cells (38-40), indicating its importance among miRNAs. miR-21 regulates biological processes, such as osteoclastogenesis, osteoclast differentiation, etc. Thus, miR-21 plays an essential role in the development of diseases, such as cancer, cardiovascular diseases and inflammation (41-43) The hsa-miR-21 gene is located on chromosome 17q23.2. Pri-miR-21 (primary transcript containing miR-21) is located within the intronic region of the tmem49 gene. Even though pri-miR-21 and tmem49 genes overlap in the same direction of transcription, pri-miR-21 is transcribed by its own promoter and is terminated with its own poly(A) tail. The pri-miR-21 transcript is subsequently processed into mature miR-21 (44,45).

The expression of miR-21 has been found to be increased in the majority of cancer types analyzed, rendering it an established oncogenic miRNA (44-54). Functional analyses in epithelial-, hepatocyte- and glial cell-derived cell lines support the regulatory role of miR-21 in cell growth, migration and invasion (46,47,55-57). Moreover, miR-21 and its associated pathways play a critical role in the pathogenesis of osteosarcoma and act as a therapeutic target for this tumor type (58). miR-21 exhibits a significantly higher expression in osteosarcoma tissues compared to adjacent normal tissues (59-61). miRNA expression has been shown to be positively associated with Enneking clinical staging and lung metastasis (62). Moreover, serum miR-21 has been reported to be a biomarker for chemosensitivity and the prognosis of human osteosarcoma (63). Additionally, circulating miR-21 levels are higher in patients with osteosarcoma than in healthy individuals. Studies have demonstrated that the detection of plasma miR-21 together with miR-143 and miR-199a-3p in patients with osteosarcoma can discriminate between the presence or absence of this tumor (62,64). The evaluation of tumor metastasis and histopathological subtype from tumors of patients with osteosarcoma has revealed a higher level of miR-21 in patients with metastatic compared to non-metastatic disease (62).

5. Exosomal miR-21 and tumor microenvironment

The tumor microenvironment (TME) differs from that of normal tissues. The TME is composed of cellular and extracellular components. The cellular components of the TME involve cancer-associated fibroblasts (CAFs), myofibroblasts, adipocytes, endothelial cells, epithelial cells and immune inflammatory cells, such as T lymphocytes, B lymphocytes, natural killer cells and natural killer T-cells, and tumor-associated macrophages (65). Cells in the TME are in constant autocrine and paracrine communication, which contributes to tumor development, progression, drug resistance and metastasis (66-70).

Exosomes provide a unique method of information transfer both locally and globally by releasing their contents into the target cell, e.g., miRNAs. By releasing exosomes, tumor cells reprogram their surroundings in the TME into a tumor-permissive or tumor-promoting environment (71-74). mir-21 present in osteosarcoma cell-derived exosomes may affect the TME (75,76), mediating the crosstalk between cancer cells, endothelial cells, immune cells, and fibroblasts in the TME to promote osteosarcoma development by i) the stimulation of tumor angiogenesis; ii) the inhibition of the immune response by acting directly on effector cells; iii) interfering with the regulation of stromal cell activation; and iv) the promotion of cancer progression by preparing the metastatic niche (Fig. 1).

miR-21 promotes cancer progression and metastasis (crosstalk among cancer cells)

Cancer is a highly heterogeneous disease. A single tumor is composed of groups of genetically clonal cells that have different growth rates, metastatic potential, invasion potential and sensitivities to chemotherapy and radiotherapy. Individual malignant tumor cells can affect the invasion and migration of surrounding tumor cells by releasing exosomal miRNAs. In contrast to exosomes released from healthy cells, exosomes derived from patients with osteosarcoma have been shown to significantly increase the adhesion, migration and viability of MG63 human osteosarcoma cells (77). Synthetic miR-143 introduced into osteosarcoma cells is released via exosomes. The delivery of exosome-contained miR-143 significantly reduces the migration of osteosarcoma cells, suggesting that miRNAs in exosomes regulate the function of cancer cells (78).

miR-21 is suggested to function as an oncogene, since it promotes the growth and development of osteosarcoma and other cancer types. In MG-63 cells, the downregulation of miR-21 results in decreased proliferation and invasion. By contrast, the elevation of miR-21 by using a mimic increases cell proliferation and invasion (79). The phosphoinositide 3-kinase (PI3K)/ R AC-α ser ine/th reon ine-protein k inase (AKT)/mammalian target of rapamycin (mTOR) signaling pathway is one of the important pathways dysregulated in osteosarcoma (80-82). miR-21 may regulate the expression of the tumor suppressor phosphatase and tensin homolog (PTEN). The downregulation or deletion of PTEN activates the PI3K/AKT (phosphoinositide 3-kinase/RAC-α serine/threonine protein kinase) signaling pathway, leading to cancer development, invasion and metastasis (83,84).

In addition to PTEN, miR-21 is suggested to target the TGF-β1 signaling pathway to promote cell proliferation in osteosarcoma. miR-21 knockdown inhibits the proliferation of osteosarcoma and promotes the expression of PTEN and TGF-β1 proteins in MG63 and U2OS human osteosarcoma cell lines (85). Moreover, it has been demonstrated that treatment with a TGF-β1 inhibitor countered the inhibitory effects of miR-21 knockdown on osteosarcoma cell proliferation (86).

Caspase-8 is a direct target of miR-21; miRNA negatively regulates the expression of caspase-8. The overexpression of miR-21 has been shown to enhance cell viability and survival, whereas it suppresses the apoptosis of the human osteosarcoma cell line, SAOS-2. In a subsequent study, miR-21 suppression was shown to increase caspase-8 expression and decrease apoptosis (60).

miR-21 is a regulator of drug resistance in osteosarcoma. The suppression of miR-21 activity has been shown to enhance the resistance of U2OS cells to cisplatin, while the ectopic expression of miR-21 in MG-63 cells reduces the resistance. Elevated miR-21 levels suppress the expression of Sprouty2. On the other hand, the ectopic expression of Sprouty2 has been shown to rescue the miR-21-mediated suppression of resistance to cisplatin, but not doxorubicin or methotrexate in osteosarcoma cells (87).

The process of metastasis is considered to be initiated in the majority, if not all, by an epithelial-to-mesenchymal transition (EMT) of the tumor cells. This allows them to migrate to and enter the vascular or lymphatic vessels, resulting in either local or distant metastasis (88,89). miR-21 has been revealed to be an important miRNA associated with cancer invasion and metastasis. The study by Yan et al (90) demonstrated that miR-21 enhanced the invasion and migration of human breast cancer MCF7 and MDA-MB-231 cells, whereas cancer cell invasion was inhibited by the knockdown of miR-21. The mechanism of metastasis regulation by miR-21 is highly complex. miR-21 targets PDCD4, tropomyosin and PTEN that are known to modulate cancer cell invasion and metastasis (91-94). Moreover, miR-21 can inhibit PTEN expression and activate AKT signaling, inducing EMT, and promoting the invasion and migration of cancer cells (95-97). Furthermore, miR-21 can trigger the interleukin (IL)-6/signal transducer and activator of transcription (STAT)3/nuclear factor (NF)-κB-mediated signaling loop for the maintenance of EMT in cancer cells (98).

miR-21 induces tumor angiogenesis (crosstalk between tumor cells and endothelial cells)

Tumor angiogenesis refers to the ability of tumor cells to recruit their own vasculature, which is critical for cancer progression. Several studies have demonstrated that exosome-encapsulated miRNAs secreted from tumor cells are able to induce angiogenesis in different cancer types (99-102). Exosomal miR-21 released by transformed lung cancer cells has been shown to induce vascular endothelial growth factor (VEGF) production and angiogenesis in nearby normal bronchial cells in a STAT3-dependent manner (99). In another study, the overexpression of miR-21 in the human prostate cancer cell line, DU145, was shown to increase the expression of Hypoxia-inducible factor (HIF)-1α and VEGF, and induce tumor angiogenesis. The miR-21-mediated activation of the AKT and extracellular signal-regulated kinase (ERK)1/2 signaling pathways enhances HIF-1α and VEGF expression (103).

miR-21 activates cancer-associated fibroblasts (CAFs; crosstalk between tumor cells and fibroblasts)

CAFs become 'activated' during the neoplastic process. CAFs are capable of accelerating the growth and promoting the invasion of tumor cells (104-109). Cancer exosomes trigger the transformation of fibroblasts through the transforming growth factor (TGF) β/Smad pathway and elicit unique effects from soluble TGFβ. The depletion of miR-21 blocks TGF-β1-induced CAF formation, whereas the overexpression of miR-21 promotes CAF induction, independent of TGF-β1. These findings clearly demonstrate that miR-21 is a critical regulator of TGF-β1 signaling induction of CAF formation (110). The conditioned medium from human lung cancer A549 cells has been shown to increase miR-21 expression and thus, through the TGF-β pathway, induce migration, CAF-like morphology and CAF markers [periostin, α-smooth muscle actin (α-SMA) and podoplanin] in human lung fibroblast MRC-5 and IMR-90 cells (111).

Exosomes from the multiple myeloma cell line, OPM2, have been showon to contain high levels of miR-21. The Co-culture of OPM2 exosomes with CAFs has also been shown to significantly increase the proliferation of and induce the CAF transformation of mesenchymal stem cells, demonstrated by the increased expression levels of fibroblast-activated protein (FAP), α-SMA and stromal-derived factor 1 (SDF-1), and the secretion of IL-6 (112). In another study, miR-21 was demonstrated to induce the metabolic alteration of CAFs and to affect the development of the human pancreatic cancer cell line, BxPC-3. Compared to normal fibroblasts, CAFs exhibited an increased miR-21 expression, glucose uptake and lactic acid production. Treatment with a miR-21 inhibitor reduced glycolysis in CAFs. The co-culture of BxPC-3 cells with CAFs treated with the miR-21 inhibitor reduced oxidative phosphorylation and the invasion of BxPC-3 cells (113).

Exosomal miR-21 suppresses the immune system (crosstalk between tumor cells and immune cells)

Exosomes secreted by tumor cells are able to inhibit the immune system by acting directly on effector cells or indirectly through their regulation (114-120). Tumor cells secrete miR-21 together with miR-29a to communicate with macrophages, eliciting a pro-inflammatory pro-metastatic response. miR-21 is a paracrine agonist of Toll-like receptor-8 (TLR-8). The activation of TLR-8 in immune cells triggers a pro-inflammatory response, that may lead to tumor growth and metastasis (121). miR-21 induces tumor-associated macrophage reprogramming through STAT3, and thereby facilitates growth, intravasation and the spread of tumor cells (122,123).

Myeloid-derived suppressor cells (MDSCs) are the major myeloid cells responsible for the immune evasion of cancer. They are composed of a heterogeneous population of immature myeloid cell progenitors, macrophage precursors, granulocytes and dendritic cells. MDSCs can promote tumor growth by the promotion of angiogenesis or the suppression of innate and adaptive immune responses. miR-21 has been shown to be upregulated in bone marrow-derived and splenic MDSCs, and increases MDSC survival and proliferation by targeting PTEN and thus, STAT3 activation (112,124).

miR-21 expression is higher in effector and memory CD8+ T-cells than in naïve CD8+ T-cells. Memory CD8+ T-cells are emerging as a key player in tumor immunosurveillance (125), and recent evidence suggests that they can control tumor growth. miR-21 negatively regulates TCR signal transduction to modulate the sensitivity of memory T-cells (118,119). Moreover, miR-21 may be involved in tumor-mediated immu-nosuppression (120,126). miR-21 from C666-1 nasopharyngeal cancer cells has been shown to activate nuclear factor 1 A-type (NF1-A) and induce IL-10 in B cells, which inhibits cytotoxic CD8+ T-cells (127).

6. Perspectives

Biogenesis of exosomal miR-21: Loading, release, and uptake. The majority of serum miRNAs is concentrated in exosomes. The loading of miRNAs into exosomes is dependent mainly on the surface molecules of exosome membranes, the endosomal sorting complex required for transport (ESCRT), and the specific binding motifs of the miRNAs themselves. A previous study demonstrated that miRNAs in exosomes are released through a ceramide-dependent secretory mechanism (128). Notably, exosomes secreted by cancer cells contain miRNAs associated with the miRNA-processing machinery RNA-induced silencing complex (RISC), and therefore, have the ability to process precursor miRNAs into mature miRNAs in a cell-independent manner, resulting in efficient and rapid reprogramming of transcriptome of the target cells (129).

Certain miRNAs are secreted selectively into exosomes. For example, the let-7 miRNA family in certain metastatic gastric cancer cell lines is secreted selectively to extracellular environment via exosomes (130). Breast cancer cell lines release the majority of miR-451 and miR-1246 selectively via exosomes, whereas both miRNAs are retained inside the non-malignant mammary epithelial cells and normal fibroblasts (131). Additionally, the TME may influence exosome release and uptake. The acidic microenvironment of tumors increases the rate of exosomal release and uptake by cancer cells (132). Furthermore, the aberrant exosome biogenesis and secretion in cancer cells, as compared with normal cells, may cause the differences in receptor recycling/degradation, plasma membrane remodeling, and the ability of endosomes to function as a signaling entity (133). In a number of types of cancer, aberrant p53 activity may result in the overexpression of the tumor suppressor-activated pathway 6 (TSAP6), through which it increases exosome production (134,135). Furthermore, heparanase, an enzyme upregulated in numerous cancer cell lines, has been shown to regulate exosome secretion in cancer cells (136).

The level of exosomal miR-21 in serum is higher in patients with osteosarcoma than in healthy individuals, which is also reflected by the high levels of miR-21 found in tumor tissue (61-64). The molecular mechanisms of exosome sorting and release have been studied extensively recently. However, the differences in miRNA content between cancer- and healthy cell-derived exosomes, the differences in exosome biogenesis between cancer and healthy cells, and the reasons why cancer cells release more exosomes with distinct content, such as miR-21, remain unclear. Additionally, whether the release of miR-21 in exosomes occurs through a distinct secretary pathway remains to be elucidated. Further investigation into the distinct exosome cargo loading mechanisms for miR-21 and other miRNAs in osteosarcoma and other cancers may aid in the identification of specific therapeutic targets related to exosome biogenesis. Further research may facilitate the use of exosomes as delivery vehicles for drugs, antigens, etc., for cancer treatment in the future.

Exosomal miR-21 as a cancer biomarker

Exosomal miR-21 is detected in human serum and plasma. Differences in miR-21 expression have been found in serum or plasma from patients with osteosarcoma and in healthy controls, supporting the role of miR-21 as a biomarker for osteosar-coma (61-64). The advantages of using exosomal miR-21 as a non-invasive biomarker are that miRNAs in blood are persistent and are highly stable against destruction by ribo-nucleases (137,138). The current methods used for exosome isolation include ultracentrifugation, size exclusion or precipitating with reagents (139). Ultracentrifugation is the commonly used method for exosome isolation. However, it is a lengthy process, which requires a large volume of samples and costly reagents and equipment. Thus, the method is impractical for clinical diagnosis. The size exclusion method uses columns to separate extracellular vesicles based on size and requires only a small amount of samples. However, the exosomes collected have heterogeneous sizes, curtailing the use for cancer diagnosis. The precipitation of exosomes with reagents not only pulls down miRNAs, but also protein aggregates that complicate the analysis for disease-specific markers.

In order to implement miR-21 as a biomarker for osteo-sarcoma, along with more effective methods for the isolation of cancer exosomes and exosomal miR-21, additional clinical trials are required to validate its use in the diagnosis of osteosarcoma and other types of cancer. Most importantly, miR-21 needs to be profiled together with other miRNAs or biomarkers specific to osteosarcoma to improve the reliability and specificity of miR-21 as a biomarker.

7. Conclusions

Exosomal miR-21 is detected in human serum and plasma. miR-21 expression in serum or plasma from patients with osteosarcoma differs from that in healthy controls, supporting the use of miR-21 as a biomarker for osteosarcoma. Exosomal miR-21 promotes cancer progression and development by mediating the crosstalk among cells in the TME. Larger-scale studies are warranted to further validate the sensitivity, specificity and applicability of circulating miR-21 as a biomarker for osteosarcoma in the future. Furthermore, extensive efforts need to be made to identify the specific cargo-loading mechanisms for exosomal miR-21. These studies will be critical for targeting miR-21 in cancer therapy to improve early-stage exosome- and miRNA-based therapeutics.

Funding

This study was supported by the National Natural Science Foundation of China (grant nos. 81672176, 81871783 and 81702582).

Availability of data and materials

Not applicable.

Authors' contributions

SW and TL wrote the manuscript. SW performed the literature search for this review article. FM and YF revised and corrected the manuscript. FM and YF assisted in the literature search for this review article and also contributed to the conception and design of the study. FM contributed to processing of the figure. TL and SH conceived and designed the study. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors confirm they have competing interests.

Acknowledgments

Not applicable.

Abbreviations:

TME

tumor microenvironment

MVEs

multivesicular endosomes

3′ UTR

3′ untranslated region

PTEN

phosphatase and tensin homolog

PI3K/AKT

phosphoinositide 3-kinase/RAC-α serine/threonine-protein kinase

EMT

epithelial-to-mesenchymal transition

VEGF

vascular endothelial growth factor

CAFs

cancer-associated fibroblasts

FAP

fibroblast-activated protein

α-SMA

α-smooth muscle actin

SDF-1

stromal-derived factor 1

TLR-8

Toll like receptor-8

MDSCs

myeloid-derived suppressor cells

ESCRT

endosomal sorting complex required for transport

RISC

RNA-induced silencing complex

TSAP6

tumor suppressor-activated pathway 6

NF1A

nuclear factor 1 A-type

References

1 

Meyers PA and Gorlick R: Osteosarcoma. Pediatr Clin North Am. 44:973–989. 1997. View Article : Google Scholar : PubMed/NCBI

2 

Ottaviani G and Jaffe N: The epidemiology of osteosarcoma. Cancer Treat Res. 152:3–13. 2009. View Article : Google Scholar

3 

Gill J, Ahluwalia MK, Geller D and Gorlick R: New targets and approaches in osteosarcoma. Pharmacol Ther. 137:89–99. 2013. View Article : Google Scholar

4 

Ostenfeld MS, Jeppesen DK, Laurberg JR, Boysen AT, Bramsen JB, Primdal-Bengtson B, Hendrix A, Lamy P, Dagnaes-Hansen F, Rasmussen MH, et al: Cellular disposal of miR23b by RAB27-dependent exosome release is linked to acquisition of metastatic properties. Cancer Res. 74:5758–5771. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Chou AJ, Geller DS and Gorlick R: Therapy for osteosarcoma: Where do we go from here? Paediatr Drugs. 10:315–327. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Sadikovic B, Yoshimoto M, Chilton-MacNeill S, Thorner P, Squire JA and Zielenska M: Identification of interactive networks of gene expression associated with osteosarcoma oncogenesis by integrated molecular profiling. Hum Mol Genet. 18:1962–1975. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Li Z, Dou P, Liu T and He S: Application of Long Noncoding RNAs in Osteosarcoma: Biomarkers and Therapeutic Targets. Cell Physiol Biochem. 42:1407–1419. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Zhou J, Liu T and Wang W: Prognostic significance of matrix metalloproteinase 9 expression in osteosarcoma: A meta-analysis of 16 studies. Medicine (Baltimore). 97:e130512018. View Article : Google Scholar

9 

Liu T, Yan Z, Liu Y, Choy E, Hornicek FJ, Mankin H and Duan Z: CRISPR-Cas9-Mediated Silencing of CD44 in Human Highly Metastatic Osteosarcoma Cells. Cell Physiol Biochem. 46:1218–1230. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Deng L, Liu T, Zhang B, Wu H, Zhao J and Chen J: Forkhead box C1 is targeted by microRNA-133b and promotes cell proliferation and migration in osteosarcoma. Exp Ther Med. 14:2823–2830. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Liu T, Li Z, Zhang Q, De Amorim Bernstein K, Lozano-Calderon S, Choy E, Hornicek FJ and Duan Z: Targeting ABCB1 (MDR1) in multi-drug resistant osteosarcoma cells using the CRISPR-Cas9 system to reverse drug resistance. Oncotarget. 7:83502–83513. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Simons M and Raposo G: Exosomes - vesicular carriers for inter-cellular communication. Curr Opin Cell Biol. 21:575–581. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Raposo G, Nijman HW, Stoorvogel W, Liejendekker R, Harding CV, Melief CJ and Geuze HJ: B lymphocytes secrete antigen-presenting vesicles. J Exp Med. 183:1161–1172. 1996. View Article : Google Scholar : PubMed/NCBI

14 

Lee TH, D'Asti E, Magnus N, Al-Nedawi K, Meehan B and Rak J: Microvesicles as mediators of intercellular communication in cancer--the emerging science of cellular 'debris'. Semin Immunopathol. 33:455–467. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Kahlert C, Melo SA, Protopopov A, Tang J, Seth S, Koch M, Zhang J, Weitz J, Chin L, Futreal A, et al: Identification of double-stranded genomic DNA spanning all chromosomes with mutated KRAS and p53 DNA in the serum exosomes of patients with pancreatic cancer. J Biol Chem. 289:3869–3875. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Villarroya-Beltri C, Baixauli F, Gutiérrez-Vázquez C, Sánchez-Madrid F and Mittelbrunn M: Sorting it out: Regulation of exosome loading. Semin Cancer Biol. 28:3–13. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Mathivanan S, Fahner CJ, Reid GE and Simpson RJ: ExoCarta 2012: Database of exosomal proteins, RNA and lipids. Nucleic Acids Res. 40(D1): D1241–D1244. 2012. View Article : Google Scholar :

18 

Mathivanan S and Simpson RJ: ExoCarta: A compendium of exosomal proteins and RNA. Proteomics. 9:4997–5000. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Pan BT, Teng K, Wu C, Adam M and Johnstone RM: Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J Cell Biol. 101:942–948. 1985. View Article : Google Scholar : PubMed/NCBI

20 

Harding C, Heuser J and Stahl P: Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J Cell Biol. 97:329–339. 1983. View Article : Google Scholar : PubMed/NCBI

21 

Keller S, Rupp C, Stoeck A, Runz S, Fogel M, Lugert S, Hager HD, Abdel-Bakky MS, Gutwein P and Altevogt P: CD24 is a marker of exosomes secreted into urine and amniotic fluid. Kidney Int. 72:1095–1102. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Taylor DD and Gercel-Taylor C: MicroRNA signatures of tumor-derived exosomes as diagnostic biomarkers of ovarian cancer. Gynecol Oncol. 110:13–21. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Gallo A, Tandon M, Alevizos I and Illei GG: The majority of microRNAs detectable in serum and saliva is concentrated in exosomes. PLoS One. 7:e306792012. View Article : Google Scholar : PubMed/NCBI

24 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Bartel DP and Chen CZ: Micromanagers of gene expression: The potentially widespread influence of metazoan microRNAs. Nat Rev Genet. 5:396–400. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Chen LT, Xu SD, Xu H, Zhang JF, Ning JF and Wang SF: MicroRNA-378 is associated with non-small cell lung cancer brain metastasis by promoting cell migration, invasion and tumor angiogenesis. Med Oncol. 29:1673–1680. 2012. View Article : Google Scholar

27 

Ivanovska I, Ball AS, Diaz RL, Magnus JF, Kibukawa M, Schelter JM, Kobayashi SV, Lim L, Burchard J, Jackson AL, et al: MicroRNAs in the miR-106b family regulate p21/CDKN1A and promote cell cycle progression. Mol Cell Biol. 28:2167–2174. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Lu Y, Thomson JM, Wong HY, Hammond SM and Hogan BL: Transgenic over-expression of the microRNA miR-17-92 cluster promotes proliferation and inhibits differentiation of lung epithelial progenitor cells. Dev Biol. 310:442–453. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Takamizawa J, Konishi H, Yanagisawa K, Tomida S, Osada H, Endoh H, Harano T, Yatabe Y, Nagino M, Nimura Y, et al: Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Res. 64:3753–3756. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Wang ZX, Bian HB, Wang JR, Cheng ZX, Wang KM and De W: Prognostic significance of serum miRNA-21 expression in human non-small cell lung cancer. J Surg Oncol. 104:847–851. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Wang X, Ling C, Bai Y and Zhao J: MicroRNA-206 is associated with invasion and metastasis of lung cancer. Anat Rec (Hoboken). 294:88–92. 2011. View Article : Google Scholar

32 

Ferracin M, Veronese A and Negrini M: Micromarkers: miRNAs in cancer diagnosis and prognosis. Expert Rev Mol Diagn. 10:297–308. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Nana-Sinkam P and Croce CM: MicroRNAs in diagnosis and prognosis in cancer: What does the future hold? Pharmacogenomics. 11:667–669. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Fan H, Lu S, Wang S and Zhang S: Identification of critical genes associated with human osteosarcoma metastasis based on integrated gene expression profiling. Mol Med Rep. 20:915–930. 2019.PubMed/NCBI

35 

Gulino R, Forte S, Parenti R, Memeo L and Gulisano M: MicroRNA and pediatric tumors: Future perspectives. Acta Histochem. 117:339–354. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Rabinowits G, Gerçel-Taylor C, Day JM, Taylor DD and Kloecker GH: Exosomal microRNA: A diagnostic marker for lung cancer. Clin Lung Cancer. 10:42–46. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Wang Y, Gao X, Wei F, Zhang X, Yu J, Zhao H, Sun Q, Yan F, Yan C, Li H, et al: Diagnostic and prognostic value of circulating miR-21 for cancer: A systematic review and meta-analysis. Gene. 533:389–397. 2014. View Article : Google Scholar

38 

Lagos-Quintana M, Rauhut R, Lendeckel W and Tuschl T: Identification of novel genes coding for small expressed RNAs. Science. 294:853–858. 2001. View Article : Google Scholar : PubMed/NCBI

39 

Lagos-Quintana M, Rauhut R, Yalcin A, Meyer J, Lendeckel W and Tuschl T: Identification of tissue-specific microRNAs from mouse. Curr Biol. 12:735–739. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Landgraf P, Rusu M, Sheridan R, Sewer A, Iovino N, Aravin A, Pfeffer S, Rice A, Kamphorst AO, Landthaler M, et al: A mammalian microRNA expression atlas based on small RNA library sequencing. Cell. 129:1401–1414. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Kumarswamy R, Volkmann I and Thum T: Regulation and function of miRNA-21 in health and disease. RNA Biol. 8:706–713. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Hu CH, Sui BD, Du FY, Shuai Y, Zheng CX, Zhao P, Yu XR and Jin Y: miR-21 deficiency inhibits osteoclast function and prevents bone loss in mice. Sci Rep. 7:431912017. View Article : Google Scholar : PubMed/NCBI

43 

Li X, Guo L, Liu Y, Su Y, Xie Y, Du J, Zhou J, Ding G, Wang H, Bai Y, et al: MicroRNA-21 promotes osteogenesis of bone marrow mesenchymal stem cells via the Smad7-Smad1/5/8-Runx2 pathway. Biochem Biophys Res Commun. 493:928–933. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Krichevsky AM and Gabriely G: miR-21: A small multi-faceted RNA. J Cell Mol Med. 13:39–53. 2009. View Article : Google Scholar : PubMed/NCBI

45 

Pan X, Wang ZX and Wang R: MicroRNA-21: A novel therapeutic target in human cancer. Cancer Biol Ther. 10:1224–1232. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Asangani IA, Rasheed SA, Nikolova DA, Leupold JH, Colburn NH, Post S and Allgayer H: MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene. 27:2128–2136. 2008. View Article : Google Scholar

47 

Frankel LB, Christoffersen NR, Jacobsen A, Lindow M, Krogh A and Lund AH: Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J Biol Chem. 283:1026–1033. 2008. View Article : Google Scholar

48 

Li T, Li D, Sha J, Sun P and Huang Y: MicroRNA-21 directly targets MARCKS and promotes apoptosis resistance and invasion in prostate cancer cells. Biochem Biophys Res Commun. 383:280–285. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Lu Z, Liu M, Stribinskis V, Klinge CM, Ramos KS, Colburn NH and Li Y: MicroRNA-21 promotes cell transformation by targeting the programmed cell death 4 gene. Oncogene. 27:4373–4379. 2008. View Article : Google Scholar : PubMed/NCBI

50 

Zhu S, Si ML, Wu H and Mo YY: MicroRNA-21 targets the tumor suppressor gene tropomyosin 1 (TPM1). J Biol Chem. 282:14328–14336. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Zhu S, Wu H, Wu F, Nie D, Sheng S and Mo YY: MicroRNA-21 targets tumor suppressor genes in invasion and metastasis. Cell Res. 18:350–359. 2008. View Article : Google Scholar : PubMed/NCBI

52 

Zheng J, Xue H, Wang T, Jiang Y, Liu B, Li J, Liu Y, Wang W, Zhang B and Sun M: miR-21 downregulates the tumor suppressor P12 CDK2AP1 and stimulates cell proliferation and invasion. J Cell Biochem. 112:872–880. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Xu J, Wu C, Che X, Wang L, Yu D, Zhang T, Huang L, Li H, Tan W, Wang C, et al: Circulating microRNAs, miR-21, miR-122, and miR-223, in patients with hepatocellular carcinoma or chronic hepatitis. Mol Carcinog. 50:136–142. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Schramedei K, Mörbt N, Pfeifer G, Läuter J, Rosolowski M, Tomm JM, von Bergen M, Horn F and Brocke-Heidrich K: MicroRNA-21 targets tumor suppressor genes ANP32A and SMARCA4. Oncogene. 30:2975–2985. 2011. View Article : Google Scholar : PubMed/NCBI

55 

Gabriely G, Wurdinger T, Kesari S, Esau CC, Burchard J, Linsley PS and Krichevsky AM: MicroRNA 21 promotes glioma invasion by targeting matrix metalloproteinase regulators. Mol Cell Biol. 28:5369–5380. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST and Patel T: MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology. 133:647–658. 2007. View Article : Google Scholar : PubMed/NCBI

57 

Si ML, Zhu S, Wu H, Lu Z, Wu F and Mo YY: miR-21-mediated tumor growth. Oncogene. 26:2799–2803. 2007. View Article : Google Scholar

58 

Sekar D, Mani P, Biruntha M, Sivagurunathan P and Karthigeyan M: Dissecting the functional role of microRNA 21 in osteosarcoma. Cancer Gene Ther. 26:179–182. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Hua Y, Jin Z, Zhou F, Zhang YQ and Zhuang Y: The expression significance of serum MiR-21 in patients with osteosarcoma and its relationship with chemosensitivity. Eur Rev Med Pharmacol Sci. 21:2989–2994. 2017.PubMed/NCBI

60 

Xu B, Xia H, Cao J, Wang Z, Yang Y and Lin Y: MicroRNA-21 Inhibits the Apoptosis of Osteosarcoma Cell Line SAOS-2 via Targeting Caspase 8. Oncol Res. 25:1161–1168. 2017. View Article : Google Scholar : PubMed/NCBI

61 

Ren X, Shen Y, Zheng S, Liu J and Jiang X: miR-21 predicts poor prognosis in patients with osteosarcoma. Br J Biomed Sci. 73:158–162. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Zhao H, Yan P, Wang J, Zhang Y, Zhang M, Wang Z, Fu Q and Liang W: Clinical significance of tumor miR-21, miR-221, miR-143, and miR-106a as biomarkers in patients with osteo-sarcoma. Int J Biol Markers. 34:184–193. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Yuan J, Chen L, Chen X, Sun W and Zhou X: Identification of serum microRNA-21 as a biomarker for chemosensitivity and prognosis in human osteosarcoma. J Int Med Res. 40:2090–2097. 2012. View Article : Google Scholar

64 

Ouyang L, Liu P, Yang S, Ye S, Xu W and Liu X: A three-plasma miRNA signature serves as novel biomarkers for osteosarcoma. Med Oncol. 30:3402013. View Article : Google Scholar

65 

Whiteside TL: The tumor microenvironment and its role in promoting tumor growth. Oncogene. 27:5904–5912. 2008. View Article : Google Scholar : PubMed/NCBI

66 

Cretu A, Brooks PC. Impact of the non-cellular tumor micro-environment on metastasis: Potential therapeutic and imaging opportunities. J Cell Physiol. 213:391–402. 2007. View Article : Google Scholar : PubMed/NCBI

67 

Hartmann S, Bhola NE and Grandis JR: HGF/Met Signaling in Head and Neck Cancer: Impact on the Tumor Microenvironment. Clin Cancer Res. 22:4005–4013. 2016. View Article : Google Scholar : PubMed/NCBI

68 

Gu F, Hu C, Tai Z, Yao C, Tian J, Zhang L, Xia Q, Gong C, Gao Y and Gao S: Tumour microenvironment-responsive lipoic acid nanoparticles for targeted delivery of docetaxel to lung cancer. Sci Rep. 6:362812016. View Article : Google Scholar : PubMed/NCBI

69 

Friedl P and Alexander S: Cancer invasion and the microenvi-ronment: Plasticity and reciprocity. Cell. 147:992–1009. 2011. View Article : Google Scholar : PubMed/NCBI

70 

Wu T, Hong Y, Jia L, Wu J, Xia J, Wang J, Hu Q and Cheng B: Modulation of IL-1β reprogrammes the tumor microenvironment to interrupt oral carcinogenesis. Sci Rep. 6:202082016. View Article : Google Scholar

71 

Maia J, Caja S, Strano Moraes MC, Couto N and Costa-Silva B: Exosome-Based Cell-Cell Communication in the Tumor Microenvironment. Front Cell Dev Biol. 6:182018. View Article : Google Scholar : PubMed/NCBI

72 

Hu C, Chen M, Jiang R, Guo Y, Wu M and Zhang X: Exosome-related tumor microenvironment. J Cancer. 9:3084–3092. 2018. View Article : Google Scholar : PubMed/NCBI

73 

Wang Z, Chen JQ, Liu JL and Tian L: Exosomes in tumor microenvironment: Novel transporters and biomarkers. J Transl Med. 14:2972016. View Article : Google Scholar : PubMed/NCBI

74 

Milane L, Singh A, Mattheolabakis G, Suresh M and Amiji MM: Exosome mediated communication within the tumor microenvi-ronment. J Control Release. 219:278–294. 2015. View Article : Google Scholar : PubMed/NCBI

75 

Raimondi L, De Luca A, Gallo A, Costa V, Russelli G, Cuscino N, Manno M, Raccosta S, Carina V, Bellavia D, et al: Osteosarcoma cell-derived exosomes affect tumor microenvironment by specific packaging of microRNAs. Carcinogenesis. Jul 10–2019.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

76 

Jerez S, Araya H, Hevia D, Irarrázaval CE, Thaler R, van Wijnen AJ and Galindo M: Extracellular vesicles from osteo-sarcoma cell lines contain miRNAs associated with cell adhesion and apoptosis. Gene. 710:246–257. 2019. View Article : Google Scholar : PubMed/NCBI

77 

Shen RK, Zhu X, Yi H, Wu CY, Chen F, Dai LQ and Lin JH: Proteomic identification of osteosarcoma-derived exosomes and their activation o f pentose phosphate pathway. Int J Clin Exp Pathol. 9:4140–4148. 2016.

78 

Shimbo K, Miyaki S, Ishitobi H, Kato Y, Kubo T, Shimose S and Ochi M: Exosome-formed synthetic microRNA-143 is transferred to osteosarcoma cells and inhibits their migration. Biochem Biophys Res Commun. 445:381–387. 2014. View Article : Google Scholar : PubMed/NCBI

79 

Lv C, Hao Y and Tu G: MicroRNA-21 promotes proliferation, invasion and suppresses apoptosis in human osteosarcoma line MG63 through PTEN/Akt pathway. Tumour Biol. 37:9333–9342. 2016. View Article : Google Scholar : PubMed/NCBI

80 

Graziano AC, Cardile V, Avola R, Vicario N, Parenti C, Salvatorelli L, Magro G and Parenti R: Wilms' tumor gene 1 silencing inhibits proliferation of human osteosarcoma MG-63 cell line by cell cycle arrest and apoptosis activation. Oncotarget. 8:13917–13931. 2017. View Article : Google Scholar : PubMed/NCBI

81 

Wang L, Tang B, Han H, Mao D, Chen J, Zeng Y and Xiong M: miR-155 Affects Osteosarcoma MG-63 Cell Autophagy Induced by Adriamycin Through Regulating PTEN-PI3K/AKT/mTOR Signaling Pathway. Cancer Biother Radiopharm. 33:32–38. 2018. View Article : Google Scholar : PubMed/NCBI

82 

Zhang J, Yu XH, Yan YG, Wang C and Wang WJ: PI3K/Akt signaling in osteosarcoma. Clin Chim Acta. 444:182–192. 2015. View Article : Google Scholar : PubMed/NCBI

83 

Wu YR, Qi HJ, Deng DF, Luo YY and Yang SL: MicroRNA-21 promotes cell proliferation, migration, and resistance to apoptosis through PTEN/PI3K/AKT signaling pathway in esophageal cancer. Tumour Biol. 37:12061–12070. 2016. View Article : Google Scholar : PubMed/NCBI

84 

Xue R, Lei S, Xia ZY, Wu Y, Meng Q, Zhan L, Su W, Liu H, Xu J, Liu Z, et al: Selective inhibition of PTEN preserves ischaemic post-conditioning cardioprotection in STZ-induced Type 1 diabetic rats: Role of the PI3K/Akt and JAK2/STAT3 pathways. Clin Sci (Lond). 130:377–392. 2016. View Article : Google Scholar

85 

Li C, Xu B, Miu X, Deng Z, Liao H and Hao L: Inhibition of miRNA-21 attenuates the proliferation and metastasis of human osteosarcoma by upregulating PTEN. Exp Ther Med. 15:1036–1040. 2018.PubMed/NCBI

86 

Hu X, Li L, Lu Y, Yu X, Chen H, Yin Q and Zhang Y: miRNA-21 inhibition inhibits osteosarcoma cell proliferation by targeting PTEN and regulating the TGF-β1 signaling pathway. Oncol Lett. 16:4337–4342. 2018.PubMed/NCBI

87 

Vanas V, Haigl B, Stockhammer V and Sutterlüty-Fall H: MicroRNA-21 Increases Proliferation and Cisplatin Sensitivity of Osteosarcoma-Derived Cells. PLoS One. 11:e01610232016. View Article : Google Scholar : PubMed/NCBI

88 

Heerboth S, Housman G, Leary M, Longacre M, Byler S, Lapinska K, Willbanks A and Sarkar S: EMT and tumor metastasis. Clin Transl Med. 4:62015. View Article : Google Scholar : PubMed/NCBI

89 

Karlsson MC, Gonzalez SF, Welin J and Fuxe J: Epithelial-mesenchymal transition in cancer metastasis through the lymphatic system. Mol Oncol. 11:781–791. 2017. View Article : Google Scholar : PubMed/NCBI

90 

Yan LX, Wu QN, Zhang Y, Li YY, Liao DZ, Hou JH, Fu J, Zeng MS, Yun JP, Wu QL, et al: Knockdown of miR-21 in human breast cancer cell lines inhibits proliferation, in vitro migration and in vivo tumor growth. Breast Cancer Res. 13:R22011. View Article : Google Scholar : PubMed/NCBI

91 

Zhao MY, Wang LM, Liu J, Huang X, Liu J and Zhang YF: MiR-21 Suppresses Anoikis through Targeting PDCD4 and PTEN in Human Esophageal Adenocarcinoma. Curr Med Sci. 38:245–251. 2018. View Article : Google Scholar : PubMed/NCBI

92 

Liao J, Liu R, Shi YJ, Yin LH and Pu YP: Exosome-shuttling microRNA-21 promotes cell migration and invasion-targeting PDCD4 in esophageal cancer. Int J Oncol. 48:2567–2579. 2016. View Article : Google Scholar : PubMed/NCBI

93 

Melnik BC: MiR-21: An environmental driver of malignant melanoma? J Transl Med. 13:2022015. View Article : Google Scholar : PubMed/NCBI

94 

Jiang LH, Ge MH, Hou XX, Cao J, Hu SS, Lu XX, Han J, Wu YC, Liu X, Zhu X, et al: miR-21 regulates tumor progression through the miR-21-PDCD4-Stat3 pathway in human salivary adenoid cystic carcinoma. Lab Invest. 95:1398–1408. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Bera A, Das F, Ghosh-Choudhury N, Kasinath BS, Abboud HE and Choudhury GG: microRNA-21-induced dissociation of PDCD4 from rictor contributes to Akt-IKKβ-mTORC1 axis to regulate renal cancer cell invasion. Exp Cell Res. 328:99–117. 2014. View Article : Google Scholar : PubMed/NCBI

96 

Zhao M, Ang L, Huang J and Wang J: MicroRNAs regulate the epithelial-mesenchymal transition and influence breast cancer invasion and metastasis. Tumour Biol. 39:10104283176916822017. View Article : Google Scholar : PubMed/NCBI

97 

Han M, Wang Y, Liu M, Bi X, Bao J, Zeng N, Zhu Z, Mo Z, Wu C and Chen X: MiR-21 regulates epithelial-mesenchymal transition phenotype and hypoxia-inducible factor-1α expression in third-sphere forming breast cancer stem cell-like cells. Cancer Sci. 103:1058–1064. 2012. View Article : Google Scholar : PubMed/NCBI

98 

De Mattos-Arruda L, Bottai G, Nuciforo PG, Di Tommaso L, Giovannetti E, Peg V, Losurdo A, Pérez-Garcia J, Masci G, Corsi F, et al: MicroRNA-21 links epithelial-to-mesenchymal transition and inflammatory signals to confer resistance to neoadjuvant trastuzumab and chemotherapy in HER2-positive breast cancer patients. Oncotarget. 6:37269–37280. 2015. View Article : Google Scholar : PubMed/NCBI

99 

Liu Y, Luo F, Wang B, Li H, Xu Y, Liu X, Shi L, Lu X, Xu W, Lu L, et al: STAT3-regulated exosomal miR-21 promotes angio-genesis and is involved in neoplastic processes of transformed human bronchial epithelial cells. Cancer Lett. 370:125–135. 2016. View Article : Google Scholar

100 

Mao G, Liu Y, Fang X, Liu Y, Fang L, Lin L, Liu X and Wang N: Tumor-derived microRNA-494 promotes angiogenesis in non-small cell lung cancer. Angiogenesis. 18:373–382. 2015. View Article : Google Scholar : PubMed/NCBI

101 

Umezu T, Tadokoro H, Azuma K, Yoshizawa S, Ohyashiki K and Ohyashiki JH: Exosomal miR-135b shed from hypoxic multiple myeloma cells enhances angiogenesis by targeting factor-inhibiting HIF-1. Blood. 124:3748–3757. 2014. View Article : Google Scholar : PubMed/NCBI

102 

Lin XJ, Fang JH, Yang XJ, Zhang C, Yuan Y, Zheng L and Zhuang SM: Hepatocellular Carcinoma Cell-Secreted Exosomal MicroRNA-210 Promotes Angiogenesis In Vitro and In Vivo. Mol Ther Nucleic Acids. 11:243–252. 2018. View Article : Google Scholar : PubMed/NCBI

103 

Liu LZ, Li C, Chen Q, Jing Y, Carpenter R, Jiang Y, Kung HF, Lai L and Jiang BH: MiR-21 induced angiogenesis through AKT and ERK activation and HIF-1α expression. PLoS One. 6:e191392011. View Article : Google Scholar

104 

Brentnall TA: Arousal of cancer-associated stromal fibroblasts: Palladin-activated fibroblasts promote tumor invasion. Cell Adhes Migr. 6:488–494. 2012. View Article : Google Scholar

105 

Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL and Weinberg RA: Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 121:335–348. 2005. View Article : Google Scholar : PubMed/NCBI

106 

Camps JL, Chang SM, Hsu TC, Freeman MR, Hong SJ, Zhau HE, von Eschenbach AC and Chung LW: Fibroblast-mediated acceleration of human epithelial tumor growth in vivo. Proc Natl Acad Sci USA. 87:75–79. 1990. View Article : Google Scholar : PubMed/NCBI

107 

Gleave M, Hsieh JT, Gao CA, von Eschenbach AC and Chung LW: Acceleration of human prostate cancer growth in vivo by factors produced by prostate and bone fibroblasts. Cancer Res. 51:3753–3761. 1991.PubMed/NCBI

108 

Kanekura T, Chen X and Kanzaki T: Basigin (CD147) is expressed on melanoma cells and induces tumor cell invasion by stimulating production of matrix metalloproteinases by fibroblasts. Int J Cancer. 99:520–528. 2002. View Article : Google Scholar : PubMed/NCBI

109 

Sameshima T, Nabeshima K, Toole BP, Yokogami K, Okada Y, Goya T and Wakisaka S: Glioma cell extracellular matrix metalloproteinase inducer (EMMPRIN) (CD147) stimulates production of membrane-type matrix metalloproteinases and activated gelatinase A in co-cultures with brain-derived fibroblasts. Cancer Lett. 157:177–184. 2000. View Article : Google Scholar : PubMed/NCBI

110 

Li Q, Zhang D, Wang Y, Sun P, Hou X, Larner J, Xiong W and Mi J: MiR-21/Smad 7 signaling determines TGF-β1-induced CAF formation. Sci Rep. 3:20382013. View Article : Google Scholar

111 

Kunita A, Morita S, Irisa TU, Goto A, Niki T, Takai D, Nakajima J and Fukayama M: MicroRNA-21 in cancer-associated fibroblasts supports lung adenocarcinoma progression. Sci Rep. 8:88382018. View Article : Google Scholar

112 

Cheng Q, Li X and Liu J, Ye Q, Chen Y, Tan S and Liu J: Multiple Myeloma-Derived Exosomes Regulate the Functions of Mesenchymal Stem Cells Partially via Modulating miR-21 and miR-146a. Stem Cells Int. 2017:90121522017. View Article : Google Scholar

113 

Mace TA, Collins AL, Wojcik SE, Croce CM, Lesinski GB and Bloomston M: Hypoxia induces the overexpression of microRNA-21 in pancreatic cancer cells. J Surg Res. 184:855–860. 2013. View Article : Google Scholar : PubMed/NCBI

114 

Muller L, Mitsuhashi M, Simms P, Gooding WE and Whiteside TL: Tumor-derived exosomes regulate expression of immune function-related genes in human T cell subsets. Sci Rep. 6:202542016. View Article : Google Scholar : PubMed/NCBI

115 

Greening DW, Gopal SK, Xu R, Simpson RJ and Chen W: Exosomes and their roles in immune regulation and cancer. Semin Cell Dev Biol. 40:72–81. 2015. View Article : Google Scholar : PubMed/NCBI

116 

Kurywchak P, Tavormina J and Kalluri R: The emerging roles of exosomes in the modulation of immune responses in cancer. Genome Med. 10:232018. View Article : Google Scholar : PubMed/NCBI

117 

Robbins PD and Morelli AE: Regulation of immune responses by extracellular vesicles. Nat Rev Immunol. 14:195–208. 2014. View Article : Google Scholar : PubMed/NCBI

118 

Wang L, He L, Zhang R, Liu X, Ren Y, Liu Z, Zhang X, Cheng W and Hua ZC: Regulation of T lymphocyte activation by microRNA-21. Mol Immunol. 59:163–171. 2014. View Article : Google Scholar : PubMed/NCBI

119 

Carissimi C, Carucci N, Colombo T, Piconese S, Azzalin G, Cipolletta E, Citarella F, Barnaba V, Macino G and Fulci V: miR-21 is a negative modulator of T-cell activation. Biochimie. 107(Pt B): 319–326. 2014. View Article : Google Scholar : PubMed/NCBI

120 

Sheedy FJ: Turning 21: Induction of miR-21 as a Key Switch in the Inflammatory Response. Front Immunol. 6:192015. View Article : Google Scholar : PubMed/NCBI

121 

Fabbri M, Paone A, Calore F, Galli R, Gaudio E, Santhanam R, Lovat F, Fadda P, Mao C, Nuovo GJ, et al: MicroRNAs bind to Toll-like receptors to induce prometastatic inflammatory response. Proc Natl Acad Sci USA. 109:E2110–E2116. 2012. View Article : Google Scholar : PubMed/NCBI

122 

Wang Z, Brandt S, Medeiros A, Wang S, Wu H, Dent A and Serezani CH: MicroRNA 21 is a homeostatic regulator of macrophage polarization and prevents prostaglandin E2-mediated M2 generation. PLoS One. 10:e01158552015. View Article : Google Scholar : PubMed/NCBI

123 

Curtale G: MiRNAs at the Crossroads between Innate Immunity and Cancer: Focus on Macrophages. Cells. 7:E122018. View Article : Google Scholar : PubMed/NCBI

124 

Li L, Zhang J, Diao W, Wang D, Wei Y, Zhang CY and Zen K: MicroRNA-155 and MicroRNA-21 promote the expansion of functional myeloid-derived suppressor cells. J Immunol. 192:1034–1043. 2014. View Article : Google Scholar : PubMed/NCBI

125 

Klebanoff CA, Gattinoni L and Restifo NP: CD8+ T-cell memory in tumor immunology and immunotherapy. Immunol Rev. 211:214–224. 2006. View Article : Google Scholar : PubMed/NCBI

126 

Cereghetti DM and Lee PP: Tumor-Derived Exosomes Contain microRNAs with Immunological Function: Implications for a Novel Immunosuppression Mechanism. MicroRNA. 2:194–204. 2014. View Article : Google Scholar : PubMed/NCBI

127 

Miao BP, Zhang RS, Li M, Fu YT, Zhao M, Liu ZG and Yang PC: Nasopharyngeal cancer-derived microRNA-21 promotes immune suppressive B cells. Cell Mol Immunol. 12:750–756. 2015. View Article : Google Scholar

128 

Kosaka N, Iguchi H, Yoshioka Y, Takeshita F, Matsuki Y and Ochiya T: Secretory mechanisms and intercellular transfer of microRNAs in living cells. J Biol Chem. 285:17442–17452. 2010. View Article : Google Scholar : PubMed/NCBI

129 

Melo SA, Sugimoto H, O'Connell JT, Kato N, Villanueva A, Vidal A, Qiu L, Vitkin E, Perelman LT, Melo CA, et al: Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell. 26:707–721. 2014. View Article : Google Scholar : PubMed/NCBI

130 

Ohshima K, Inoue K, Fujiwara A, Hatakeyama K, Kanto K, Watanabe Y, Muramatsu K, Fukuda Y, Ogura S, Yamaguchi K, et al: Let-7 microRNA family is selectively secreted into the extracellular environment via exosomes in a metastatic gastric cancer cell line. PLoS One. 5:e132472010. View Article : Google Scholar : PubMed/NCBI

131 

Pigati L, Yaddanapudi SC, Iyengar R, Kim DJ, Hearn SA, Danforth D, Hastings ML and Duelli DM: Selective release of microRNA species from normal and malignant mammary epithelial cells. PLoS One. 5:e135152010. View Article : Google Scholar : PubMed/NCBI

132 

Boussadia Z, Lamberti J, Mattei F, Pizzi E, Puglisi R, Zanetti C, Pasquini L, Fratini F, Fantozzi L, Felicetti F, et al: Acidic micro-environment plays a key role in human melanoma progression through a sustained exosome mediated transfer of clinically relevant metastatic molecules. J Exp Clin Cancer Res. 37:2452018. View Article : Google Scholar

133 

Hessvik NP and Llorente A: Current knowledge on exosome biogenesis and release. Cell Mol Life Sci. 75:193–208. 2018. View Article : Google Scholar :

134 

Lespagnol A, Duflaut D, Beekman C, Blanc L, Fiucci G, Marine JC, Vidal M, Amson R and Telerman A: Exosome secretion, including the DNA damage-induced p53-dependent secretory pathway, is severely compromised in TSAP6/Steap3-null mice. Cell Death Differ. 15:1723–1733. 2008. View Article : Google Scholar : PubMed/NCBI

135 

Yu X, Harris SL and Levine AJ: The regulation of exosome secretion: A novel function of the p53 protein. Cancer Res. 66:4795–4801. 2006. View Article : Google Scholar : PubMed/NCBI

136 

Thompson CA, Purushothaman A, Ramani VC, Vlodavsky I and Sanderson RD: Heparanase regulates secretion, composition, and function of tumor cell-derived exosomes. J Biol Chem. 288:10093–10099. 2013. View Article : Google Scholar : PubMed/NCBI

137 

Wang K, Zhang S, Marzolf B, Troisch P, Brightman A, Hu Z, Hood LE and Galas DJ: Circulating microRNAs, potential biomarkers for drug-induced liver injury. Proc Natl Acad Sci USA. 106:4402–4407. 2009. View Article : Google Scholar : PubMed/NCBI

138 

Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O'Briant KC, Allen A, et al: Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA. 105:10513–10518. 2008. View Article : Google Scholar : PubMed/NCBI

139 

Greening DW, Xu R, Ji H, Tauro BJ and Simpson RJ: A protocol for exosome isolation and characterization: Evaluation of ultra-centrifugation, density-gradient separation, and immunoaffinity capture methods. Methods Mol Biol. 1295:179–209. 2015. View Article : Google Scholar

Related Articles

Journal Cover

May-2020
Volume 56 Issue 5

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang S, Ma F, Feng Y, Liu T and He S: Role of exosomal miR‑21 in the tumor microenvironment and osteosarcoma tumorigenesis and progression (Review). Int J Oncol 56: 1055-1063, 2020
APA
Wang, S., Ma, F., Feng, Y., Liu, T., & He, S. (2020). Role of exosomal miR‑21 in the tumor microenvironment and osteosarcoma tumorigenesis and progression (Review). International Journal of Oncology, 56, 1055-1063. https://doi.org/10.3892/ijo.2020.4992
MLA
Wang, S., Ma, F., Feng, Y., Liu, T., He, S."Role of exosomal miR‑21 in the tumor microenvironment and osteosarcoma tumorigenesis and progression (Review)". International Journal of Oncology 56.5 (2020): 1055-1063.
Chicago
Wang, S., Ma, F., Feng, Y., Liu, T., He, S."Role of exosomal miR‑21 in the tumor microenvironment and osteosarcoma tumorigenesis and progression (Review)". International Journal of Oncology 56, no. 5 (2020): 1055-1063. https://doi.org/10.3892/ijo.2020.4992