Next Article in Journal
Association between Vitamin D Deficiency and High Serum Levels of Small Dense LDL in Middle-Aged Adults
Next Article in Special Issue
Regulation of MRP4 Expression by circHIPK3 via Sponging miR-124-3p/miR-4524-5p in Hepatocellular Carcinoma
Previous Article in Journal
Degeneration of Aortic Valves in a Bioreactor System with Pulsatile Flow
Previous Article in Special Issue
Circulating microRNAs Related to Bone Metabolism in HIV-Associated Bone Loss
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of MicroRNAs in Human Osteosarcoma: Future Perspectives

1
Department of Animal Production and Health, Public Veterinary Health and Food Science and Technology, Faculty of Veterinary Medicine, Universidad Cardenal Herrera-CEU, CEU Universities, 46115 Alfara del Patriarca, Spain
2
Atlantia Veterinary Hospital, 44200 Nantes, France
*
Author to whom correspondence should be addressed.
Biomedicines 2021, 9(5), 463; https://doi.org/10.3390/biomedicines9050463
Submission received: 14 March 2021 / Revised: 21 April 2021 / Accepted: 21 April 2021 / Published: 23 April 2021

Abstract

:
Osteosarcoma (OS) is a rare form of cancer with high death rate but is one of the most frequent forms of bone cancer in children and adolescents. MiRNAs are small endogenous RNAs that regulate gene expression post-transcriptionally. The discovery of miRNAs could allow us to obtain an earlier diagnosis, predict prognosis and chemoresistance, and lead to the discovery of new treatments in different types of tumors, including OS. Despite the fact that there is currently only one clinical trial being carried out on a single miRNA for solid tumors, it is very probable that the number of clinical trials including miRNAs as prognostic and diagnostic biomarkers, as well as potential therapeutic targets, will increase in the near future. This review summarizes the different miRNAs related to OS and their possible therapeutic application.

1. Introduction

Osteosarcoma (OS) is the most common primary bone tumor in children and adolescents. Although advances made in chemotherapy, radiation therapy, and surgery have improved survival rates in different cancers, little to no progress has been seen in recent years for OS [1,2]. Unfortunately, in osteosarcoma, 15–20% of patients admitted for treatment, already have an advanced form of the disease with metastases, that are most commonly found in the lungs [3,4,5]. These lung metastases are what cause the death of the patient in 30–40% of cases [6]. In patients without lung metastases, the 5-year survival rate is 58–75% [7]. This prognosis is determined by the rate of occurrence of metastasis and the appearance of chemoresistance [6]. Given the aggressiveness and high mortality rates of OS, early detection is key to a successful treatment. Recent studies have shown that OS is becoming increasingly resistant to chemotherapeutic agents. Therefore, understanding the underlying molecular mechanisms causing the resistance, and the development of novel and more successful treatments, has become a necessity. In fact, Basile et al. (2020) published a meta-analysis in which they found different prognostic factors in patients under the age of 40, enabling an accurate prediction of the risk a patient has of developing lung metastasis. Optimizing the treatment of patients, based on these different prognostic factors, could play an important role in increasing OS survival rates. [8]. In recent years, microRNAs (miRNAs) have been presented as possible diagnostic and prognostic biomarkers, as modulators of chemoresistance and as a plausible new treatment option in the fight against different tumors, including OS.

2. Human Osteosarcoma

Osteosarcoma (OS) is a high-grade malignant bone tumor composed of mesenchymal stem cells (malignant osteoblasts). It is characterized by the deposition of an immature osteoid matrix resulting in the formation of immature woven bone. The incidence of this tumor varies according to race, age, sex, and other factors. It is more frequent in males, children aged 10–14, and in African Americans, suggesting that osteosarcoma may have genetic predisposition [9]. Hereditary disorders such as retinoblastoma, the Li-Fraumeni syndrome, Rothmund–Thomson syndrome, Bloom’s syndrome and Werner syndrome [10], Paget’s disease, chromosomal abnormalities, ionizing radiation, and alkylating agents [11], are risk factors related to the progress of the disease. Current treatment consists of preoperative chemotherapy to shrink the primary tumor and its posterior resection. For the first step, a combination of drugs is commonly used in order to avoid chemoresistance and increase the degree of tumor necrosis. The most common chemotherapeutic drugs include DOX (doxorubicine), cisplatin, ifosfamide, and a high-dose of methotrexate associated with leucovorin calcium as a rescue agent [12]. Three to four weeks after completion of the latter treatment, surgical resection of the tumor is carried out, and 2 weeks after surgery postoperative chemotherapy is administered [13]. Currently, other treatments are in the clinical trial phase, and their results are not yet known. Treatments, currently in phases II and III of clinical trials include, inhibitors of platelet-derived growth factor (PDFG), RANKL and bisphosphonate, vascular endothelial growth factor (VEGF), mTOR, immune checkpoints, antagonists of insulin growth factor 1 receptor (IGF-1R), Tyrosine kinase Src, human epidermal growth factor receptor (HER2/Neu) blockers, cytokines, immunomodulating agents, dendritic cell peptide vaccines, and chimeric antigen receptor (CAR) T cells [14]. Despite advances in treatments, the 5-year survival rate in patients with metastases—which is usually around 50% of patients initiating treatment—has not improved in the last decade and remains below 30% [15,16]. In addition, the number of patients that develop drug resistances are significant. This highlights the importance of finding early diagnostic and prognostic markers and identifying new therapeutic targets in the fight against OS [3].
In the last decade, a new group of molecules known as miRNAs, able to act as genetic biomarkers in numerous diseases including OS, have emerged [17]. They are non-coding RNA molecules of 20–24 base pairs in length, which are unable to code for proteins. They are phylogenetically conserved and have a crucial role in the regulation of gene expression and cellular processes [18,19,20,21,22].

3. MicroRNAs: Biogenesis and Biological Functions

3.1. Biogenesis of miRNAs

Biogenesis of miRNAs begins in the cell nucleus, where miRNAs are transcribed by RNA polymerase II or III as primary transcripts (pri-mRNA) to specific genes. Later, the enzyme Drosha and its cofactor DGCR8/Pasha cut this pri-miRNA to form a pre-miRNA (70–90 nucleotides long in the shape of a stem-loop). The nuclear transport receptor Exportin 5 (XPO5), that plays an important role in various cancers, then actively transports the pre-miRNA from the nucleus to the cytoplasm. In the cytoplasm, the pre-miRNA is cut by a Dicer enzyme (regulated by XPO5), to form a mature and short double-stranded miRNA molecule and, after the degradation of one of the two chains, a single strand of miRNA is incorporated into the RISC (RNA-induced silencing complex) protein complex. The RISC protein complex is responsible for directing the silencing of messenger RNA and regulating post transcriptional gene expression, via the inhibition of protein translation or destabilization of target transcripts [23,24,25,26,27,28,29,30,31] (Figure 1).

3.2. Biological Functions of miRNAs

MiRNAs are distributed in the intragenic and intergenic regions of the genome. miRNA genes are observed in defined units or clusters [32], containing two or more adjacent sequences with a common transcriptional triggering mechanism. These clusters are involved in different biological functions, as regulators of biological events such as cellular homeostasis, and their aberrant expression has been associated with pathophysiological events [33].
Biological events regulated by miRNAs include differentiation and development, nervous system regulation, immunity, viral infection, DNA repair, cell junctions and cell to cell communication, cellular reprogramming, and metabolism, among others [33]. For example, miR-17/92 is highly expressed in embryonic stem cells and is related to skeletal development, lung morphogenesis, cardiomyocyte and epithelial proliferation, pre-eclampsia and reduction of trophoblast differentiation, and early embryonic lethality [34,35,36,37]. Furthermore, this cluster is associated with B-cell apoptosis, [38] T-cell proliferation [39], DNA repair, post-natal neovascularization [40], mitochondrial metabolism [41], and DNA repair via deacetylation activity of sirtuin 1 (SIRT1) [42]. Other miRNA clusters with different functions include miR-106b/25, related to spermatogonial differentiation [43], generation of new neurons [44], early embryonic lethality [37], coronary artery disease [45], or control of Ca2+ accumulation in the mitochondrial matrix [46]. Kabekkodu et al. (2018) published a relevant review of these miRNA clusters among others, and their roles in biological functions [33].
Regarding cancer, the main hallmarks are cell cycle control, apoptosis and senescence, autophagy, cell proliferation and growth, angiogenesis, and metastasis. miRNA clusters play a crucial role in these steps, and their role and type can differ depending on the type of cancer. More specifically, the miR-17/92 and miR106b/25 clusters are related to proliferation, tumor initiation, metastasis, senescence, cell cycle regulation, apoptosis, and autophagy [47,48,49,50,51,52,53,54,55,56,57]. Other miRNA clusters, their pathways, and related cancer hallmarks are shown in Figure 2.
Indeed, some of these clusters are associated with cell cycle progression, migration, and metastasis in osteosarcomas. More specifically, miR-17/92 is found to be abnormally regulated in human OS cell lines and is linked to cell cycle progression [58] and activation of migration and metastasis via the downregulation of the Quaking (QKI) and β-catenin pathways [59]. In fact, patients with a high expression of miR-17/92 presented poor recurrence-free and overall survival rates [60].

3.3. MiRNAs and Human Osteosarcoma

Many studies reflect the important role that miRNAs play in the molecular regulation, the appearance and the progression of human OS. Some of these miRNAs act as tumor suppressors, whilst others act as oncogenes [61]. Different miRNAs will act on a diverse range of molecular targets and pathways. Different tumor suppressing miRNAs and their molecular targets in human OS are shown in Table 1.
The presence of onco-miRNAs affecting different targets in human osteosarcoma is demonstrated in Table 2.
Interestingly, miRNAs can act as both tumor suppressors and as onco-miRNAs depending on the target that they act upon. For example, miR-23a can act as tumor suppressor in osteosarcomas, via the inhibition of the special AT-rich-binding protein 1 (SATB1), a gene related to chromatin structure regulation [62]. This finding is supported by Wang et al. who suggests that its role as a tumor suppressor could serve as a potential therapeutic target to fight osteosarcoma. MiR-23a is frequently found to be downregulated in osteosarcoma specimens and cell lines in vitro, which contributes to the aggressiveness of this cancer [63]. Indeed, an experiment carried out by He et al. in an in vivo mice model showed that the downregulation of miR-23a activates the transcription of its target mRNAs, RUNX2 and CXCL12, which play an essential role in OS cellular growth, migration, and invasion [64]. On the contrary, other studies have shown miR-23a acting as an onco-miRNA, and in this case, its downregulation could effectively reduce migration and invasion of osteosarcoma cell lines. As a matter of fact, miR-23a specifically targets the 3′-untranslational regions of PTEN (phosphatase and tensin homolog), lowering its expression. This enhances migration and invasion of osteosarcoma cell lines and might serve as a risk factor for cancer patients [65]. Similar results have been found for miR-20a and miR-107. Depending on which target they bind to, these miRNAs are able to act as both onco-genes or as tumor suppressors. Indeed, the increased expression of miR-20a can reduce colony formation and tumor growth in OS cells, by targeting and suppressing TAK1 expression [66], whereas when this miRNA binds to early growth factor 2 (EGF2) mRNA, it promotes OS cell proliferation and cell cycle [67]. Furthermore, miR-20a seems to participate in the regulation of OS chemoresistance by modulating the activities of the MAPK/ERK and cAMP/PKA signaling pathways [68]. Meanwhile, miR-107 is able to activate MEK/ERK and NF-κB signaling pathways via downregulation of tropomyosin 1 (TPM1). This promotes cell viability, migration, and invasion in human OS cells [69]. Conversely, in vitro studies have demonstrated that miR-107 can inhibit the development of OS via the Wnt/β-catenin signaling pathways, by downregulating Dickkopf-1 (Dkk-1) (Wnt inhibitory factor) [70,71].

4. Possible Applications of miRNAs in Human Osteosarcoma

As shown in the previous section, miRNAs have the capacity to act as tumor suppressors and as onco-miRNAs in human osteosarcomas, by acting on different signaling pathways. In fact, more than 177 different miRNAs are found to be differentially expressed in osteosarcoma cell lines [72], and could serve as prognostic, diagnostic, and chemoresistance biomarkers. MiRNAs could also act as possible therapeutic targets to treat human OS. In the following section, the possible uses of certain relevant miRNAs are detailed.

4.1. MiRNAs as Potential Biomarkers

One of the most studied miRNAs in osteosarcoma is miR-9. Recently, Wu et al. published a meta-analysis and concluded that a high expression of miR-9 was associated with poor prognosis [73]. Patients with highly expressed levels of miR-9 in osteosarcoma tissues present bigger tumor size, more advanced clinical stages, more frequent distant metastasis, and lower survival rates [74]. The effect that miR-9 has on OS progression seems to be related to it targeting p16 and mediating the activation of the ERK/p38/JNK pathways [75]. Interestingly, the high expression of miR-9, occurs not only in OS tissues, but also in the serum of patients [76]. Similar results have been obtained for the miR-29 family (miR-29a, miR-29b, and miR-29c) [77]. In this case, the underlying mechanism seems to be that miR-29 inhibits the expression of PTEN, thus promoting OS cell proliferation, migration, and metastasis formation [78]. With regards to metastasis occurrence, high circulating levels of miR-21 in patients’ blood sera have been positively correlated with the presence of metastasis and lower 5-year survival rates [79,80]. MiR-95 promotes OS growth by targeting the sodium channel epithelial 1α subunit (SCNN1A) and hepatoma-derived growth factor (HDGF) [81,82]. Other circulating miRNAs found in patients’ sera include miR-95 and miR-194. The higher expression of miR-95 and the lower expression of miR-194 in patients’ sera, contributes to higher metastasis formation and leads to a poorer prognosis in patients with OS [83,84]. These results suggest that altered serum levels of miR-9, miR-21, miR-29, miR-95, and miR-194 have great potential to serve as novel, non-invasive prognostic or diagnostic biomarkers.
Some miRNAs are related to p53, a known oncogene found in many cancers. One of these miRNAs is miR-34a, who’s downregulation in human OS tissues is associated with an unfavorable prognosis [85]. In fact, Xi et al. demonstrated that the downregulation of miR-34 in the tissues of OS patients, inhibited apoptosis through targeting regulated transforming growth factor-β-induced factor homeobox 2s’ (TGIF2) expression [86]. Another miRNA whose expression is lower in OS tissues is miR-126. This miRNA is able to downregulate metalloprotease 9 (ADAM-9), a protease that plays a role in promoting tumorigenesis, cell invasion, and metastasis [87]. Therefore, lower levels of miR-126 could potentially be associated with faster tumor progression, more advanced cancer stages, and shorter overall survival times [88].
MultimiRNA signature for prediction of prognosis of OS patients, has been studied, and three miRNAs (miR-153, miR-212, and miR-591) were used to generate a classified prognosis and prediction signature for OS patients [89]. It was found that miR-153 could potentially act as a tumor suppressor, as high levels of the latter decrease SMAD2, SMAD3, EGFR, and IGFBP-3 expressions, which in turn negatively regulates TGF-β [90]. On the contrary, miR-212 has been seen to inhibit apoptosis and promotes cell proliferation, by upregulating the hedgehog signaling pathway [91]. As for miR-591, although its presence in osteosarcoma has not yet been studied, its function as a tumor suppressor in breast cancer has been suggested, via the inhibition of the Hippo-YAP/TAZ signaling pathway [92]. In addition, miR-591 upregulation has been seen to confer resistance to paclitaxel, a medication used to treat a number of cancers including ovarian cancer. To date however, no research has been carried out on the effects of miR-591 on cisplatin or methotrexate resistance (the most common drugs used to treat OS) [93].

4.2. MiRNAs and OS Chemoresistance

One of the most important steps in the human OS is the patient’s response to treatment. The presence of certain miRNAs seems to affect tumor cell chemoresistance. Curiously, high levels of the miR-29 family, which are related to a poor prognosis (see Section 4.1), sensitizes tumor cells against methotrexate and increases the effect of carbon ion radiotherapy [94,95]. The combined use of cisplatin in addition to methotrexate is common when trying to treat osteosarcoma. In a study carried out by Jiang L et al., these two chemotherapy drugs were seen to promote apoptosis in cells that overexpressed miR-126, but not in the silenced osteosarcoma cells [96]. This suggests that the differential expression of miRNAs may play an important role in chemotherapy resistance. Finally, high levels of miR-34a-5p were seen to affect the expression of the tyrosine kinase receptor CD117, thus promoting osteosarcoma’s multidrug resistance via the mTOR pathway [97,98].

4.3. MiRNAs as Targets for OS Therapy

Different roles of miR-1 have been studied in diseases such as cardiac diseases, sclerosis, pulmonary arterial hypertension, non-alcoholic fatty liver disease, and cerebral palsy [99,100,101,102]. Furthermore, miR-1 has been related to different types of human cancers such as medulloblastoma, breast cancer, colorectal cancer, and lung cancer [103,104,105,106]. In osteosarcoma, some studies reveal its potential as a therapeutic target; for example, miR-1 is able to suppress growth, proliferation, migration, and invasion of OS cells. In an experiment carried out by Fujii et al., overexpression of miR-1 downregulated PAX3 (and increased p21 levels), and induced arrest in the G0/G1 phase both in vitro and in vivo [107]. In addition, in vitro studies have shown that miR-1 inhibits the protein expression of vascular endothelial growth factor A (VEGFA), a necessary factor for the vascularization of tumors [108]. These results suggest that miR-1 could be a potential therapeutic target, via the p21 or VEGFA pathways. Regarding the latter pathway, miR-134 (a miRNA of the 14q32 locus) is capable of decreasing VEGFA and VEGFR1 expression in vivo [109], and inhibiting osteosarcoma angiogenesis and proliferation. Moreover, when this microRNA binds to the 3′-UTRs of metalloproteinases 1 and 3 (MMP1 and MMP3), it reduces tumor invasion and metastasis [110]. Therefore, miR-134 could also act as a possible therapeutic target. Other members of the 14q32 locus, such as miR-382, miR-369-3p and miR-544, suppress the expression of the c-MYC (oncogene) in human OS cells [111]. Studies in patient OS cells have demonstrated that another MMP, more precisely MMP16, is upregulated by miR-145 [112]. However, MMP16 is not miR-154′s only target. Other molecules, such as ROCK1, VEGF, and HDAC4 are also targets of this miRNA [113,114,115]. High levels of miR-145 are seen to inhibit cell proliferation, invasion, and metastasis in OS. A recent meta-analysis carried out by Xu et al. [116] showed that the level at which miR-145 was expressed in different tumors was correlated with patient prognosis, however this was not shown in patients with OS. Nevertheless, other studies related the downregulation of miR-145 with advanced tumor progression and poor prognosis in OS [117]. Another microRNA, miR-21, has been put forward as alternative possible therapeutic target. Indeed, miR-21 suppresses PTEN expression, and induces apoptosis in human osteosarcoma cells [118].
There are a large number of studies providing relevant information about the possible functions of microRNAs as potential biomarkers for prognosis, diagnosis and resistance to chemotherapy or as possible therapeutic targets in different cancers. Despite the good potential OS has, as a target of future clinical trials based on miRNA therapy, the truth is that there are actually only very few clinical trials based on miRNAs in this cancer to date. Possible explanations for this are that as a rare form cancer, there are only very few patients and therefore the recruitment potential for clinical trials is lower than it is in other cancer types. In addition, obtaining miRNAs from a bone microenvironment requires complicated procedures. Finally, the complexity of the human genome makes it difficult to interpret the results obtained from miRNA trials. We are still uncertain of the effects each miRNA may exert on the rest of the genome, and resistance mechanisms are likely. As a matter of fact, some clinical trials for solid tumors based on miRNA have been carried out. One of them was based on the therapeutic molecule MRX34, whose target is miR-34a. The results of MRX34 treatment with dexamethasone premedication did show some clinical activity without serious toxicity. However, some patients presented severe cytokine release syndrome, and the trial was terminated [119]. The final results of this clinical trial are not published yet. Another clinical trial, based on miR-16 (a tumor suppressor miRNA), has been carried out in lung cancer and mesothelioma, and included 27 patients. Although the safety of the trial was acceptable, twenty of patients died during the clinical trial due to tumor progression [120]. The results found in these different clinical trials indicate the amount of knowledge that still needs to be acquired on miRNAs before they can be used as biomarkers or as cancer therapies, including data on their safety and effectiveness.
Currently, there are a total of 334 clinical trials addressing miRNAs as possible biomarkers or therapeutic targets in different types of cancer. However, only one of them is for osteosarcoma (NCT01190943). This clinical trial is studying DNA biomarkers, including miRNAs, in tissue samples from patients with osteosarcoma. Other clinical trials, based on different miRNAs in the hope of treating further pathologies are currently being carried out. More specifically, currently, two clinical trials with therapeutic molecules targeting miRNAs in other pathologies are being carried out, and they are showing promising results. These molecules target miR-122 for the treatment of the hepatitis C viral infection and are in phases 1 and 2 of clinical trials [121,122].

5. Conclusions

OSs are a rare form of cancer with high death rates in humans, mainly children. The discovery of miRNAs marked a new era in molecular biology and their presence can give valuable information about the physiopathology of osteosarcoma. The relationship between different miRNAs and patient prognosis, cancer diagnosis, cancer progression, chemotherapy resistance, and their use as possible new drug targets have been demonstrated. However, there is currently only one clinical trial related to the inhibition of miRNA in solid tumors; this miRNA is miR-34a. It seems that one of the issues is the delivery system of the therapies. Another issue is the localized delivery of miRNA therapeutics. The hope is that in the near future these drawbacks will be overcome, and anti-cancer therapy based on miRNAs will be a reality, especially for solid tumors such as OS, with poor prognosis and high mortality rates.

Author Contributions

writing—original draft preparation, O.G. and L.L.; writing—review and editing, O.G. and L.L.; supervision, L.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We are grateful to the Veterinary Medicine Faculty of Universidad Cardenal Herrera-CEU.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Anderson, M.E. Update on Survival in Osteosarcoma. Orthop. Clin. N. Am. 2016, 47, 283–292. [Google Scholar] [CrossRef] [PubMed]
  2. Smrke, A.; Anderson, P.; Gulia, A.; Gennatas, S.; Huang, P.; Jones, R. Future Directions in the Treatment of Osteosarcoma. Cells 2021, 10, 172. [Google Scholar] [CrossRef] [PubMed]
  3. Kushlinskii, N.E.; Fridman, M.V.; Braga, E.A. Long Non-Coding RNAs as Competitive Endogenous RNAs in Osteosarcoma. Mol. Biol. 2020, 54, 776–801. [Google Scholar] [CrossRef]
  4. Bielack, S.S.; Kempf-Bielack, B.; Branscheid, D.; Carrle, D.; Friedel, G.; Helmke, K.; Kevric, M.; Jundt, G.; Kühne, T.; Maas, R.; et al. Second and Subsequent Recurrences of Osteosarcoma: Presentation, Treatment, and Outcomes of 249 Consecutive Cooperative Osteosarcoma Study Group Patients. J. Clin. Oncol. 2009, 27, 557–565. [Google Scholar] [CrossRef]
  5. Aljubran, A.H.; Griffin, A.; Pintilie, M.; Blackstein, M. Osteosarcoma in adolescents and adults: Survival analysis with and without lung metastases. Ann. Oncol. 2009, 20, 1136–1141. [Google Scholar] [CrossRef] [PubMed]
  6. Sasaki, R.; Osaki, M.; Okada, F. MicroRNA-Based Diagnosis and Treatment of Metastatic Human Osteosarcoma. Cancers 2019, 11, 553. [Google Scholar] [CrossRef] [Green Version]
  7. Isakoff, M.S.; Bielack, S.S.; Meltzer, P.S.; Gorlick, R. Osteosarcoma: Current Treatment and a Collaborative Pathway to Success. J. Clin. Oncol. 2015, 33, 3029–3035. [Google Scholar] [CrossRef] [Green Version]
  8. Basile, P.; Greengard, E.; Weigel, B.; Spector, L. Prognostic Factors for Development of Subsequent Metastases in Localized Osteosarcoma: A Systematic Review and Identification of Literature Gaps. Sarcoma 2020, 2020, 1–7. [Google Scholar] [CrossRef] [Green Version]
  9. Sadykova, L.R.; Ntekim, A.I.; Muyangwa-Semenova, M.; Rutland, C.S.; Jeyapalan, J.N.; Blatt, N.; Rizvanov, A.A. Epidemiology and Risk Factors of Osteosarcoma. Cancer Investig. 2020, 38, 259–269. [Google Scholar] [CrossRef]
  10. Rickel, K.; Fang, F.; Tao, J. Molecular genetics of osteosarcoma. Bone 2017, 102, 69–79. [Google Scholar] [CrossRef]
  11. Jafari, F.; Javdansirat, S.; Sanaie, S.; Naseri, A.; Shamekh, A.; Rostamzadeh, D.; Dolati, S. Osteosarcoma: A comprehensive review of management and treatment strategies. Ann. Diagn. Pathol. 2020, 49, 151654. [Google Scholar] [CrossRef]
  12. Wittig, J.C.; Bickels, J.; Priebat, D.; Jelinek, J.; Kellar-Graney, K.; Shmookler, B.; Malawer, M.M. Osteosarcoma: A multidisciplinary approach to diagnosis and treatment. Am. Fam. Physician 2002, 65, 1123–1132. [Google Scholar] [PubMed]
  13. Chindamo, G.; Sapino, S.; Peira, E.; Chirio, D.; Gonzalez, M.C.; Gallarate, M. Bone Diseases: Current Approach and Future Perspectives in Drug Delivery Systems for Bone Targeted Therapeutics. Nanomaterials 2020, 10, 875. [Google Scholar] [CrossRef] [PubMed]
  14. Fernandes, I.; Melo-Alvim, C.; Lopes-Brás, R.; Esperança-Martins, M.; Costa, L. Osteosarcoma Pathogenesis Leads the Way to New Target Treatments. Int. J. Mol. Sci. 2021, 22, 813. [Google Scholar] [CrossRef]
  15. He, X.; Gao, Z.; Xu, H.; Zhang, Z.; Fu, P. A meta-analysis of randomized control trials of surgical methods with osteosarcoma outcomes. J. Orthop. Surg. Res. 2017, 12, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Zhu, L.; McManus, M.M.; Hughes, D.P.M. Understanding the Biology of Bone Sarcoma from Early Initiating Events through Late Events in Metastasis and Disease Progression. Front. Oncol. 2013, 3, 230. [Google Scholar] [CrossRef] [Green Version]
  17. Gourbault, O.; Llobat, L. MicroRNAs as Biomarkers in Canine Osteosarcoma: A New Future? Vet. Sci. 2020, 7, 146. [Google Scholar] [CrossRef]
  18. Chen, X. A MicroRNA as a Translational Repressor of APETALA2 in Arabidopsis Flower Development. Science 2004, 303, 2022–2025. [Google Scholar] [CrossRef] [Green Version]
  19. Doench, J.G. Specificity of microRNA target selection in translational repression. Genes Dev. 2004, 18, 504–511. [Google Scholar] [CrossRef] [Green Version]
  20. Heneghan, H.M.; Miller, N.; Kerin, M.J. MiRNAs as biomarkers and therapeutic targets in cancer. Curr. Opin. Pharmacol. 2010, 10, 543–550. [Google Scholar] [CrossRef]
  21. Macfarlane, L.-A.; Murphy, P.R. MicroRNA: Biogenesis, Function and Role in Cancer. Curr. Genom. 2010, 11, 537–561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Kim, V.N.; Han, J.; Siomi, M.C. Biogenesis of small RNAs in animals. Nat. Rev. Mol. Cell Biol. 2009, 10, 126–139. [Google Scholar] [CrossRef] [PubMed]
  23. Ballarino, M.; Pagano, F.; Girardi, E.; Morlando, M.; Cacchiarelli, D.; Marchioni, M.; Proudfoot, N.J.; Bozzoni, I. Coupled RNA Processing and Transcription of Intergenic Primary MicroRNAs. Mol. Cell. Biol. 2009, 29, 5632–5638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Han, J.; Lee, Y.; Yeom, K.-H.; Kim, Y.-K.; Jin, H.; Kim, V.N. The Drosha-DGCR8 complex in primary microRNA processing. Genes Dev. 2004, 18, 3016–3027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Lee, Y.; Ahn, C.; Han, J.; Choi, H.; Kim, J.; Yim, J.; Lee, J.; Provost, P.; Rådmark, O.; Kim, S.; et al. The nuclear RNase III Drosha initiates microRNA processing. Nat. Cell Biol. 2003, 425, 415–419. [Google Scholar] [CrossRef] [PubMed]
  26. Morlando, M.; Ballarino, M.; Gromak, N.; Pagano, F.; Bozzoni, I.; Proudfoot, N.J. Primary MicroRNA Transcripts are Processed Co-Transcriptionally. Nat. Struct. Mol. Biol. 2008, 15, 902–909. [Google Scholar] [CrossRef]
  27. Bohnsack, M.T.; Czaplinski, K.; Görlich, D. Exportin 5 is a RanGTP-dependent dsRNA-binding protein that mediates nuclear export of pre-miRNAs. RNA 2004, 10, 185–191. [Google Scholar] [CrossRef] [Green Version]
  28. Lund, E.; Güttinger, S.; Calado, A.; Dahlberg, J.E.; Kutay, U. Nuclear Export of MicroRNA Precursors. Science 2004, 303, 95–98. [Google Scholar] [CrossRef] [Green Version]
  29. Yi, R.; Qin, Y.; Macara, I.G.; Cullen, B.R. Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev. 2003, 17, 3011–3016. [Google Scholar] [CrossRef] [Green Version]
  30. Hutvágner, G.; McLachlan, J.; Pasquinelli, A.E.; Bálint, É.; Tuschl, T.; Zamore, P.D. A Cellular Function for the RNA-Interference Enzyme Dicer in the Maturation of the let-7 Small Temporal RNA. Science 2001, 293, 834–838. [Google Scholar] [CrossRef] [Green Version]
  31. Ketting, R.F.; Fischer, S.E.; Bernstein, E.; Sijen, T.; Hannon, G.J.; Plasterk, R.H. Dicer functions in RNA interference and in synthesis of small RNA involved in developmental timing in C. elegans. Genes Dev. 2001, 15, 2654–2659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Altuvia, Y. Clustering and conservation patterns of human microRNAs. Nucleic Acids Res. 2005, 33, 2697–2706. [Google Scholar] [CrossRef] [PubMed]
  33. Kabekkodu, S.P.; Shukla, V.; Varghese, V.K.; Souza, J.D.; Chakrabarty, S.; Satyamoorthy, K. Clustered miRNAs and their role in biological functions and diseases. Biol. Rev. 2018, 93, 1955–1986. [Google Scholar] [CrossRef]
  34. Mogilyansky, E.; Rigoutsos, I. The miR-17/92 cluster: A comprehensive update on its genomics, genetics, functions and increasingly important and numerous roles in health and disease. Cell Death Differ. 2013, 20, 1603–1614. [Google Scholar] [CrossRef]
  35. Yan, S.; Jiao, K. Functions of miRNAs during Mammalian Heart Development. Int. J. Mol. Sci. 2016, 17, 789. [Google Scholar] [CrossRef]
  36. Kumar, P.; Luo, Y.; Tudela, C.; Alexander, J.M.; Mendelson, C.R. The c-Myc-Regulated MicroRNA-17∼92 (miR-17∼92) and miR-106a∼363 Clusters Target hCYP19A1 and hGCM1 To Inhibit Human Trophoblast Differentiation. Mol. Cell. Biol. 2013, 33, 1782–1796. [Google Scholar] [CrossRef] [Green Version]
  37. Ventura, A.; Young, A.G.; Winslow, M.M.; Lintault, L.; Meissner, A.; Erkeland, S.J.; Newman, J.; Bronson, R.T.; Crowley, D.; Stone, J.R.; et al. Targeted Deletion Reveals Essential and Overlapping Functions of the miR-17∼92 Family of miRNA Clusters. Cell 2008, 132, 875–886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Xiao, C.; Srinivasan, L.; Calado, D.P.; Patterson, H.C.; Zhang, B.; Wang, J.; Henderson, J.M.; Kutok, J.L.; Rajewsky, K. Lymphoproliferative disease and autoimmunity in mice with increased miR-17-92 expression in lymphocytes. Nat. Immunol. 2008, 9, 405–414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Baumjohann, D.; Kageyama, R.; Clingan, J.M.; Morar, M.M.; Patel, S.; De Kouchkovsky, D.; Bannard, O.; Bluestone, J.A.; Matloubian, M.; Ansel, K.M.; et al. The microRNA cluster miR-17∼92 promotes TFH cell differentiation and represses subset-inappropriate gene expression. Nat. Immunol. 2013, 14, 840–848. [Google Scholar] [CrossRef]
  40. Doebele, C.; Bonauer, A.; Fischer, A.; Scholz, A.; Reiss, Y.; Urbich, C.; Hofmann, W.-K.; Zeiher, A.M.; Dimmeler, S. Members of the microRNA-17-92 cluster exhibit a cell-intrinsic antiangiogenic function in endothelial cells. Blood 2010, 115, 4944–4950. [Google Scholar] [CrossRef] [Green Version]
  41. Hajarnis, S.; Lakhia, R.; Yheskel, M.; Williams, D.; Sorourian, M.; Liu, X.; Aboudehen, K.; Zhang, S.; Kersjes, K.; Galasso, R.; et al. microRNA-17 family promotes polycystic kidney disease progression through modulation of mitochondrial metabolism. Nat. Commun. 2017, 8, 14395. [Google Scholar] [CrossRef] [PubMed]
  42. Xie, L.; Huang, R.; Liu, S.; Wu, W.; Su, A.; Li, R.; Liu, X.; Lei, Y.; Sun, H.; Liu, X.; et al. A positive feedback loop of SIRT1 and miR17HG promotes the repair of DNA double-stranded breaks. Cell Cycle 2019, 18, 2110–2123. [Google Scholar] [CrossRef] [PubMed]
  43. Tong, M.H.; Mitchell, D.A.; McGowan, S.D.; Evanoff, R.; Griswold, M.D. Two miRNA Clusters, Mir-17-92 (Mirc1) and Mir-106b-25 (Mirc3), Are Involved in the Regulation of Spermatogonial Differentiation in Mice1. Biol. Reprod. 2012, 86, 72. [Google Scholar] [CrossRef] [PubMed]
  44. Brett, J.O.; Renault, V.M.; Rafalski, V.A.; Webb, A.E.; Brunet, A. The microRNA cluster miR-106b~25 regulates adult neural stem/progenitor cell proliferation and neuronal differentiation. Aging 2011, 3, 108–124. [Google Scholar] [CrossRef] [Green Version]
  45. Bronze-Da-Rocha, E. MicroRNAs Expression Profiles in Cardiovascular Diseases. BioMed Res. Int. 2014, 2014, 1–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Marchi, S.; Pinton, P. Mitochondrial calcium uniporter, MiRNA and cancer. Commun. Integr. Biol. 2013, 6, e23818. [Google Scholar] [CrossRef] [Green Version]
  47. Conkrite, K.; Sundby, M.; Mukai, S.; Thomson, J.M.; Mu, D.; Hammond, S.M.; MacPherson, D. miR-17 92 cooperates with RB pathway mutations to promote retinoblastoma. Genes Dev. 2011, 25, 1734–1745. [Google Scholar] [CrossRef] [Green Version]
  48. Jiang, H.; Wang, P.; Wang, Q.; Wang, B.; Mu, J.; Zhuang, X.; Zhang, L.; Yan, J.; Miller, D.; Zhang, H.-G. Quantitatively Controlling Expression of miR-17∼92 Determines Colon Tumor Progression in a Mouse Tumor Model. Am. J. Pathol. 2014, 184, 1355–1368. [Google Scholar] [CrossRef] [Green Version]
  49. Hong, L.; Lai, M.; Chen, M.; Xie, C.; Liao, R.; Kang, Y.J.; Xiao, C.; Hu, W.Y.; Han, J.; Sun, P. The miR-17-92 Cluster of MicroRNAs Confers Tumorigenicity by Inhibiting Oncogene-Induced Senescence. Cancer Res. 2010, 70, 8547–8557. [Google Scholar] [CrossRef] [Green Version]
  50. Brockway, S.; Zeleznik-Le, N.J. WEE1 is a validated target of the microRNA miR-17-92 cluster in leukemia. Cancer Genet. 2015, 208, 279–287. [Google Scholar] [CrossRef] [Green Version]
  51. Gits, C.; Van Kuijk, P.F.; Jonkers, M.B.E.; Boersma, A.W.M.; Van Ijcken, W.F.; Wozniak, A.; Sciot, R.; Rutkowski, P.; Schoffski, P.; Taguchi, T.; et al. MiR-17-92 and miR-221/222 cluster members target KIT and ETV1 in human gastrointestinal stromal tumours. Br. J. Cancer 2013, 109, 1625–1635. [Google Scholar] [CrossRef]
  52. Urtasun, R.; Elizalde, M.; Azkona, M.; Latasa, M.U.; García-Irigoyen, O.; Uriarte, I.; Fernández-Barrena, M.G.; Vicent, S.; Alonso, M.M.; Muntané, J.; et al. Splicing regulator SLU7 preserves survival of hepatocellular carcinoma cells and other solid tumors via oncogenic miR-17-92 cluster expression. Oncogene 2016, 35, 4719–4729. [Google Scholar] [CrossRef] [PubMed]
  53. Li, Y.; Tan, W.; Neo, T.W.; Aung, M.O.; Wasser, S.; Lim, S.G.; Tan, T.M. Role of themiR-106b-25microRNA cluster in hepatocellular carcinoma. Cancer Sci. 2009, 100, 1234–1242. [Google Scholar] [CrossRef] [PubMed]
  54. Smith, A.L.; Iwanaga, R.; Drasin, D.J.; Micalizzi, D.S.; Vartuli, R.L.; Tan, A.-C.; Ford, H.L. The miR-106b-25 cluster targets Smad7, activates TGF-β signaling, and induces EMT and tumor initiating cell characteristics downstream of Six1 in human breast cancer. Oncogene 2012, 31, 5162–5171. [Google Scholar] [CrossRef] [Green Version]
  55. Savita, U.; Karunagaran, D. MicroRNA-106b-25 cluster targets β-TRCP2, increases the expression of Snail and enhances cell migration and invasion in H1299 (non small cell lung cancer) cells. Biochem. Biophys. Res. Commun. 2013, 434, 841–847. [Google Scholar] [CrossRef]
  56. Zhou, Y.; Hu, Y.; Yang, M.; Jat, P.; Li, K.; Lombardo, Y.; Xiong, D.; Coombes, R.C.; Raguz, S.; Yagüe, E. The miR-106b∼25 cluster promotes bypass of doxorubicin-induced senescence and increase in motility and invasion by targeting the E-cadherin transcriptional activator EP300. Cell Death Differ. 2014, 21, 462–474. [Google Scholar] [CrossRef] [Green Version]
  57. Kan, T.; Sato, F.; Ito, T.; Matsumura, N.; David, S.; Cheng, Y.; Agarwal, R.; Paun, B.C.; Jin, Z.; Olaru, A.V.; et al. The miR-106b-25 Polycistron, Activated by Genomic Amplification, Functions as an Oncogene by Suppressing p21 and Bim. Gastroenterology 2009, 136, 1689–1700. [Google Scholar] [CrossRef] [Green Version]
  58. Baumhoer, D.; Zillmer, S.; Unger, K.; Rosemann, M.; Atkinson, M.J.; Irmler, M.; Beckers, J.; Siggelkow, H.; Von Luettichau, I.; Jundt, G.; et al. MicroRNA profiling with correlation to gene expression revealed the oncogenic miR-17-92 cluster to be up-regulated in osteosarcoma. Cancer Genet. 2012, 205, 212–219. [Google Scholar] [CrossRef]
  59. Yang, H.; Peng, Z.; Liang, M.; Zhang, Y.; Wang, Y.; Huang, T.; Jiang, Y.; Jiang, B.; Wang, Y. The miR-17-92 cluster/QKI2/β-catenin axis promotes osteosarcoma progression. Oncotarget 2018, 9, 25285–25293. [Google Scholar] [CrossRef] [PubMed]
  60. Li, X.; Yang, H.; Tian, Q.; Liu, Y.; Weng, Y. Upregulation of microRNA-17-92 cluster associates with tumor progression and prognosis in osteosarcoma. Neoplasma 2014, 61, 453–460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Viera, G.M.; Salomao, K.B.; De Sousa, G.R.; Baroni, M.; Delsin, L.E.A.; Pezuk, J.A.; Brassesco, M.S. miRNA signatures in childhood sarcomas and their clinical implications. Clin. Transl. Oncol. 2019, 21, 1583–1623. [Google Scholar] [CrossRef] [PubMed]
  62. Cai, S.; Han, H.-J.; Kohwi-Shigematsu, T. Tissue-specific nuclear architecture and gene expession regulated by SATB1. Nat. Genet. 2003, 34, 42–51. [Google Scholar] [CrossRef] [PubMed]
  63. Wang, G.; Li, B.; Fu, Y.; He, M.; Wang, J.; Shen, P.; Bai, L. miR-23a suppresses proliferation of osteosarcoma cells by targeting SATB1. Tumor Biol. 2015, 36, 4715–4721. [Google Scholar] [CrossRef] [PubMed]
  64. He, Y.; Meng, C.; Shao, Z.; Wang, H.; Yang, S. MiR-23a Functions as a Tumor Suppressor in Osteosarcoma. Cell. Physiol. Biochem. 2014, 34, 1485–1496. [Google Scholar] [CrossRef] [PubMed]
  65. Tian, K.; Di, R.; Wang, L. MicroRNA-23a enhances migration and invasion through PTEN in osteosarcoma. Cancer Gene Ther. 2015, 22, 351–359. [Google Scholar] [CrossRef] [Green Version]
  66. Yuan, G.; Zhao, Y.; Wu, D.; Gao, C.; Jiao, Z. miRNA-20a upregulates TAK1 and increases proliferation in osteosarcoma cells. Futur. Oncol. 2018, 14, 461–469. [Google Scholar] [CrossRef]
  67. Zhuo, W.; Ge, W.; Meng, G.; Jia, S.; Zhou, X.; Liu, J. MicroRNA-20a promotes the proliferation and cell cycle of human osteosarcoma cells by suppressing early growth response 2 expression. Mol. Med. Rep. 2015, 12, 4989–4994. [Google Scholar] [CrossRef] [Green Version]
  68. Pu, Y.; Yi, Q.; Zhao, F.; Wang, H.; Cai, W.; Cai, S. MiR-20a-5p represses multi-drug resistance in osteosarcoma by targeting the KIF26B gene. Cancer Cell Int. 2016, 16, 1–15. [Google Scholar] [CrossRef] [Green Version]
  69. Jiang, R.; Zhang, C.; Liu, G.; Gu, R.; Wu, H. MicroRNA-107 Promotes Proliferation, Migration, and Invasion of Osteosarcoma Cells by Targeting Tropomyosin 1. Oncol. Res. Featur. Preclin. Clin. Cancer Ther. 2017, 25, 1409–1419. [Google Scholar] [CrossRef]
  70. Rossini, M.; Gatti, D.; Adami, S. Involvement of WNT/β-catenin Signaling in the Treatment of Osteoporosis. Calcif. Tissue Int. 2013, 93, 121–132. [Google Scholar] [CrossRef] [PubMed]
  71. Zhang, Z.C.; Liu, J.X.; Shao, Z.W.; Pu, F.F.; Wang, B.C.; Wu, Q.; Zhang, Y.K.; Zeng, X.L.; Guo, X.D.; Yang, S.H.; et al. In vitro effect of microRNA-107 targeting Dkk-1 by regulation of Wnt/β-catenin signaling pathway in osteosarcoma. Medicine 2017, 96, e7245. [Google Scholar] [CrossRef]
  72. Namløs, H.M.; Meza-Zepeda, L.A.; Barøy, T.; Østensen, I.H.G.; Kresse, S.H.; Kuijjer, M.L.; Serra, M.; Bürger, H.; Cleton-Jansen, A.M.; Myklebost, O. Modulation of the Osteosarcoma Expression Phenotype by MicroRNAs. PLoS ONE 2012, 7, e48086. [Google Scholar] [CrossRef] [Green Version]
  73. Wu, F.; Jiang, X.; Wang, Q.; Lu, Q.; He, F.; Li, J.; Li, X.; Jin, M.; Xu, J. The impact of miR-9 in osteosarcoma. Medicine 2020, 99, e21902. [Google Scholar] [CrossRef] [PubMed]
  74. Xu, S.H.; Yang, Y.L.; Han, S.M.; Wu, Z.H. MicroRNA-9 expression is a prognostic biomarker in patients with osteosarcoma. World J. Surg. Oncol. 2014, 12, 195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Gao, S.; Wang, J.; Tian, S.; Luo, J. miR‑9 depletion suppresses the proliferation of osteosarcoma cells by targeting p16. Int. J. Oncol. 2019, 54, 1921–1932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Fei, D.; Li, Y.; Zhao, D.; Zhao, K.; Dai, L.; Gao, Z. Serum miR-9 as a prognostic biomarker in patients with osteosarcoma. J. Int. Med. Res. 2014, 42, 932–937. [Google Scholar] [CrossRef] [PubMed]
  77. Hong, Q.; Fang, J.; Pang, Y.; Zheng, J. Prognostic value of the microRNA-29 family in patients with primary osteosarcomas. Med. Oncol. 2014, 31, 1–8. [Google Scholar] [CrossRef] [PubMed]
  78. Liu, Q.; Geng, P.; Shi, L.; Wang, Q.; Wang, P. miR-29 promotes osteosarcoma cell proliferation and migration by targeting PTEN. Oncol. Lett. 2018, 17, 883–890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Ouyang, L.; Liu, P.; Yang, S.; Ye, S.; Xu, W.; Liu, X. A three-plasma miRNA signature serves as novel biomarkers for osteosarcoma. Med. Oncol. 2012, 30, 340. [Google Scholar] [CrossRef] [PubMed]
  80. Hua, Y.; Jin, Z.; Zhou, F.; Zhang, Y.-Q.; Zhuang, Y. The expression significance of serum MiR-21 in patients with osteosarcoma and its relationship with chemosensitivity. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 2989–2994. [Google Scholar]
  81. Geng, Y.; Zhao, S.; Jia, Y.; Xia, G.; Li, H.; Fang, Z.; Zhang, Q.; Tian, R. miR‑95 promotes osteosarcoma growth by targeting SCNN1A. Oncol. Rep. 2020, 43, 1429–1436. [Google Scholar] [CrossRef]
  82. Liu, X.; Ma, W.; Ma, J.; Xiao, L.; Hao, D. Upregulation of miR‑95-3p inhibits growth of osteosarcoma by targeting HDGF. Pathol. Res. Pr. 2019, 215, 152492. [Google Scholar] [CrossRef]
  83. Niu, J.; Sun, Y.; Guo, Q.; Niu, D.; Liu, B. Serum miR-95-3p is a diagnostic and prognostic marker for osteosarcoma. SpringerPlus 2016, 5, 1–6. [Google Scholar] [CrossRef] [Green Version]
  84. Shi, L.; Xie, C.; Zhu, J.; Chen, X. Downregulation of serum miR-194 predicts poor prognosis in osteosarcoma patients. Ann. Diagn. Pathol. 2020, 46, 151488. [Google Scholar] [CrossRef]
  85. Wang, Y.; Jia, L.S.; Yuan, W.; Wu, Z.; Wang, H.B.; Xu, T.; Sun, J.C.; Cheng, K.F.; Shi, J.G. Low miR-34a and miR-192 are associated with unfavorable prognosis in patients suffering from osteosarcoma. Am. J. Transl. Res. 2015, 7, 111–119. [Google Scholar]
  86. Xi, L.; Zhang, Y.; Kong, S.; Liang, W. miR-34 inhibits growth and promotes apoptosis of osteosarcoma in nude mice through targetly regulating TGIF2 expression. Biosci. Rep. 2018, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Jiang, L.; He, A.; Zhang, Q.; Tao, C. miR-126 inhibits cell growth, invasion, and migration of osteosarcoma cells by downregulating ADAM-9. Tumor Biol. 2014, 35, 12645–12654. [Google Scholar] [CrossRef] [PubMed]
  88. Liu, W.; Zhao, Z.Y.; Shi, L.; Yuan, W.D. Tissue microRNA-126 expression level predicts outcome in human osteosarcoma. Diagn. Pathol. 2015, 10, 116. [Google Scholar] [CrossRef] [Green Version]
  89. Luo, X.; Tang, J.; Xuan, H.; Liu, J.; Li, X. Identification and Validation of a Potent Multi-miRNA Signature for Prediction of Prognosis of Osteosarcoma Patients. Med. Sci. Monit. 2020, 26, e919272-1. [Google Scholar] [CrossRef] [PubMed]
  90. Niu, G.; Li, B.; Sun, L.; An, C. MicroRNA-153 Inhibits Osteosarcoma Cells Proliferation and Invasion by Targeting TGF-β2. PLoS ONE 2015, 10, e0119225. [Google Scholar] [CrossRef] [PubMed]
  91. Yi, T.; Yi, X.; Lv, X.; Liang, R.; Zan, N.; Su, X. MiR-212 promotes proliferation and inhibits apoptosis of osteosarcoma cells via regulating hedgehog signaling pathway. J. BUON 2020, 25, 2086–2091. [Google Scholar]
  92. Huang, X.; Tang, F.; Weng, Z.; Zhou, M.; Zhang, Q. MiR-591 functions as tumor suppressor in breast cancer by targeting TCF4 and inhibits Hippo-YAP/TAZ signaling pathway. Cancer Cell Int. 2019, 19, 1–9. [Google Scholar] [CrossRef]
  93. Huh, J.H.; Kim, T.H.; Kim, K.; Song, J.A.; Jung, Y.J.; Jeong, J.Y.; Lee, M.J.; Kim, Y.K.; Lee, D.H.; An, H.J. Dysregulation of miR-106a and miR-591 confers paclitaxel resistance to ovarian cancer. Br. J. Cancer 2013, 109, 452–461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Xu, W.; Li, Z.; Zhu, X.; Xu, R.; Xu, Y. miR-29 Family Inhibits Resistance to Methotrexate and Promotes Cell Apoptosis by Targeting COL3A1 and MCL1 in Osteosarcoma. Med. Sci. Monit. 2018, 24, 8812–8821. [Google Scholar] [CrossRef] [PubMed]
  95. Kim, E.H.; Kim, J.Y.; Kim, M.S.; Vares, G.; Ohno, T.; Takahashi, A.; Uzawa, A.; Seo, S.J.; Sai, S. Molecular mechanisms underlying the enhancement of carbon ion beam radiosensitivity of osteosarcoma cells by miR-29b. Am. J. Cancer Res. 2020, 10, 4357–4371. [Google Scholar] [PubMed]
  96. Jiang, L.; He, A.; He, X.; Tao, C. MicroRNA-126 enhances the sensitivity of osteosarcoma cells to cisplatin and methotrexate. Oncol. Lett. 2015, 10, 3769–3778. [Google Scholar] [CrossRef] [Green Version]
  97. Pu, Y.; Zhao, F.; Wang, H.; Cai, W.; Gao, J.; Li, Y.; Cai, S. MiR-34a-5p promotes the multi-drug resistance of osteosarcoma by targeting the CD117 gene. Oncotarget 2016, 7, 28420–28434. [Google Scholar] [CrossRef] [Green Version]
  98. Zhou, Y.; Zhao, R.H.; Tseng, K.F.; Li, K.P.; Lu, Z.G.; Liu, Y.; Han, K.; Gan, Z.H.; Lin, S.C.; Hu, H.Y.; et al. Sirolimus induces apoptosis and reverses multidrug resistance in human osteosarcoma cells in vitro via increasing microRNA-34b expression. Acta Pharmacol. Sin. 2016, 37, 519–529. [Google Scholar] [CrossRef] [Green Version]
  99. Li, E.Y.; Zhao, P.J.; Jian, J.; Yin, B.Q.; Sun, Z.Y.; Xu, C.X.; Tang, Y.C.; Wu, H. Vitamin B1 and B12 mitigates neuron apoptosis in cerebral palsy by augmenting BDNF expression through MALAT1/miR-1 axis. Cell Cycle 2019, 18, 2849–2859. [Google Scholar] [CrossRef] [PubMed]
  100. Jiang, F.; Chen, Q.; Wang, W.; Ling, Y.; Yan, Y.; Xia, P. Hepatocyte-derived extracellular vesicles promote endothelial inflammation and atherogenesis via microRNA-1. J. Hepatol. 2020, 72, 156–166. [Google Scholar] [CrossRef]
  101. Lv, L.; Zheng, N.; Zhang, L.; Li, R.; Li, Y.; Yang, R.; Li, C.; Fang, R.; Shabanova, A.; Li, X.; et al. Metformin ameliorates cardiac conduction delay by regulating microRNA-1 in mice. Eur. J. Pharmacol. 2020, 881, 173131. [Google Scholar] [CrossRef] [PubMed]
  102. Mondejar-Parreño, G.; Callejo, M.; Barreira, B.; Morales-Cano, D.; Esquivel-Ruiz, S.; Moreno, L.; Cogolludo, A.; Perez-Vizcaino, F. miR-1 is increased in pulmonary hypertension and downregulates Kv1.5 channels in rat pulmonary arteries. J. Physiol. 2018, 597, 1185–1197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Peng, J.; Yuan, C.; Wu, Z.; Wang, Y.; Yin, W.; Lin, Y.; Zhou, L.; Lu, J. Upregulation of microRNA‑1 inhibits proliferation and metastasis of breast cancer. Mol. Med. Rep. 2020, 22, 454–464. [Google Scholar] [CrossRef]
  104. Wu, Y.; Pu, N.; Su, W.; Yang, X.; Xing, C. Downregulation of miR-1 in colorectal cancer promotes radioresistance and aggressive phenotypes. J. Cancer 2020, 11, 4832–4840. [Google Scholar] [CrossRef]
  105. Zhang, J.; Li, N.; Fu, J.; Zhou, W. Long noncoding RNA HOTAIR promotes medulloblastoma growth, migration and invasion by sponging miR-1/miR-206 and targeting YY1. Biomed. Pharmacother. 2020, 124, 109887. [Google Scholar] [CrossRef] [PubMed]
  106. Sheervalilou, R.; Lotfi, H.; Shirvaliloo, M.; Sharifi, A.; Nazemiyeh, M.; Zarghami, N. Circulating MiR-10b, MiR-1 and MiR-30a Expression Profiles in Lung Cancer: Possible Correlation with Clinico-Pathologic Characteristics and Lung Cancer Detection. Int. J. Mol. Cell Med. 2019, 8, 118–129. [Google Scholar]
  107. Fujii, R.; Osaka, E.; Sato, K.; Tokuhashi, Y. MiR-1 Suppresses Proliferation of Osteosarcoma Cells by Up-regulating p21 via PAX3. Cancer Genom. Proteom. 2019, 16, 71–79. [Google Scholar] [CrossRef] [Green Version]
  108. Niu, J.; Sun, Y.; Guo, Q.; Niu, D.; Liu, B. miR-1 Inhibits Cell Growth, Migration, and Invasion by Targeting VEGFA in Osteosarcoma Cells. Dis. Markers 2016, 2016, 1–8. [Google Scholar] [CrossRef] [Green Version]
  109. Zhang, L.; Lv, Z.; Xu, J.; Chen, C.; Ge, Q.; Li, P.; Wei, D.; Wu, Z.; Sun, X. Micro RNA -134 inhibits osteosarcoma angiogenesis and proliferation by targeting the VEGFA/VEGFR 1 pathway. FEBS J. 2018, 285, 1359–1371. [Google Scholar] [CrossRef] [Green Version]
  110. Chen, C.; Zhang, L.; Jiao, Y.; Zhou, Y.; Ge, Q.; Li, P.; Sun, X.; Lv, Z. miR-134 inhibits osteosarcoma cell invasion and metastasis through targeting MMP 1 and MMP 3 in vitro and in vivo. FEBS Lett. 2019, 593, 1089–1101. [Google Scholar] [CrossRef]
  111. Thayanithy, V.; Sarver, A.L.; Kartha, R.V.; Li, L.; Angstadt, A.Y.; Breen, M.; Steer, C.J.; Modiano, J.F.; Subramanian, S. Perturbation of 14q32 miRNAs-cMYC gene network in osteosarcoma. Bone 2012, 50, 171–181. [Google Scholar] [CrossRef] [Green Version]
  112. Chen, B.; Huang, Z.; Zhang, Y.; Chen, Y.; Li, Z. MicroRNA-145 Suppresses Osteosarcoma Metastasis via Targeting MMP16. Cell. Physiol. Biochem. 2015, 37, 2183–2193. [Google Scholar] [CrossRef]
  113. Wu, G.; Yu, W.; Zhang, M.; Yin, R.; Wu, Y.; Liu, Q. MicroRNA-145-3p suppresses proliferation and promotes apotosis and autophagy of osteosarcoma cell by targeting HDAC4. Artif. Cells Nanomed. Biotechnol. 2018, 46, 579–586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Li, E.; Zhang, J.; Yuan, T.; Ma, B. miR-145 inhibits osteosarcoma cells proliferation and invasion by targeting ROCK1. Tumor Biol. 2014, 35, 7645–7650. [Google Scholar] [CrossRef] [PubMed]
  115. Fan, L.; Wu, Q.; Xing, X.; Wei, Y.; Shao, Z. MicroRNA-145 targets vascular endothelial growth factor and inhibits invasion and metastasis of osteosarcoma cells. Acta Biochim. Biophys. Sin. 2012, 44, 407–414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Xu, L.; Zhang, Y.; Tang, J.; Wang, P.; Li, L.; Yan, X.; Zheng, X.; Ren, S.; Zhang, M.; Xu, M. The Prognostic Value and Regulatory Mechanisms of microRNA-145 in Various Tumors: A Systematic Review and Meta-analysis of 50 Studies. Cancer Epidemiol. Biomarkers Prev. 2019, 28, 867–881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Zhou, Z.; Tang, M.; Lin, L.; Cai, H.; Tang, J. MicroRNA-145 downregulation associates with advanced tumor progression and poor prognosis in patients suffering osteosarcoma. Oncol. Targets Ther. 2013, 6, 833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Yang, J.; Zou, Y.; Jiang, D. Honokiol suppresses proliferation and induces apoptosis via regulation of the miR‑21/PTEN/PI3K/AKT signaling pathway in human osteosarcoma cells. Int. J. Mol. Med. 2018, 41, 1845–1854. [Google Scholar] [CrossRef] [PubMed]
  119. Hong, D.S.; Kang, Y.-K.; Borad, M.; Sachdev, J.; Ejadi, S.; Lim, H.Y.; Brenner, A.J.; Park, K.; Lee, J.-L.; Kim, T.-Y.; et al. Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours. Br. J. Cancer 2020, 122, 1630–1637. [Google Scholar] [CrossRef] [PubMed]
  120. van Zandwijk, N.; Pavlakis, N.; Kao, S.C.; Linton, A.; Boyer, M.J.; Clarke, S.; Huynh, Y.; Chrzanowska, A.; Fulham, M.J.; Bailey, D.L.; et al. Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: A first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 2017, 18, 1386–1396. [Google Scholar] [CrossRef]
  121. van der Ree, M.H.; de Vree, J.M.; Stelma, F.; Willemse, S.; van der Valk, M.; Rietdijk, S.; Molenkamp, R.; Schinkel, J.; van Nuenen, A.C.; Beuers, U.; et al. Safety, tolerability, and antiviral effect of RG-101 in patients with chronic hepatitis C: A phase 1B, double-blind, randomised controlled trial. Lancet 2017, 389, 709–717. [Google Scholar] [CrossRef]
  122. Van Der Ree, M.H.; Van Der Meer, A.J.; Van Nuenen, A.C.; De Bruijne, J.; Ottosen, S.; Janssen, H.L.; Kootstra, N.A.; Reesink, H.W. Miravirsen dosing in chronic hepatitis C patients results in decreased microRNA-122 levels without affecting other microRNAs in plasma. Aliment. Pharmacol. Ther. 2016, 43, 102–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Diagram of miRNA biogenesis. In the nucleus, miRNA that are transcribed by RNA polymerase II or III as primary transcripts (pri-miRNA). The enzyme Drosha and its cofactor DGCR8/Pasha then cut the pri-miRNA molecule to form a pre-miRNA. This pre-miRNA is actively transported from the nucleus to the cytoplasm by the nuclear transport receptor Exportin 5 (XPO5) in a Ran-GTP (guanosine triphosphate) protein dependent manner by XPO5. In the cytoplasm, the pre-miRNA is cut by a second enzyme called Dicer, to form a mature and short double-stranded miRNA molecule. The miRNA duplex unwinds and is incorporated into the RISC protein complex [17].
Figure 1. Diagram of miRNA biogenesis. In the nucleus, miRNA that are transcribed by RNA polymerase II or III as primary transcripts (pri-miRNA). The enzyme Drosha and its cofactor DGCR8/Pasha then cut the pri-miRNA molecule to form a pre-miRNA. This pre-miRNA is actively transported from the nucleus to the cytoplasm by the nuclear transport receptor Exportin 5 (XPO5) in a Ran-GTP (guanosine triphosphate) protein dependent manner by XPO5. In the cytoplasm, the pre-miRNA is cut by a second enzyme called Dicer, to form a mature and short double-stranded miRNA molecule. The miRNA duplex unwinds and is incorporated into the RISC protein complex [17].
Biomedicines 09 00463 g001
Figure 2. MiRNA clusters, their pathways and associated cancer hallmarks. AKT, AKT serine/threonine kinase; BH3, Bcl-2 homology 3 domain; BIM, Bcl-2-like protein 11; EP300, E1A-asosciated protein p300; ETV1, Ets variant gene 1; KIT, proto-oncogene tyrosine-protein kinase; MET, MET proto-oncogene receptor tyrosine kinase; mTOR, mammalian target of rapamycin complex 1; p21, cyclin dependent kinase inhibitor 1A; PLCG1, phospholipase c gamma 1; PSAP, prosaposin; p53, tumor protein p53; PTEN, phosphatase and tensin homolog; RB1, RB transcriptional corepressor 1; RHO, ROCK Rho-associated protein kinase; SLU7, pre-mRNA splicing factor SLU7; SMAD2, mothers against dpp homolog 2; TRCP2, F-box and WD repeat domain containing 11; TGF, transforming growth factor; WEE1, Wee1A kinase; Wnt, wingless-type mmtv integration site family.
Figure 2. MiRNA clusters, their pathways and associated cancer hallmarks. AKT, AKT serine/threonine kinase; BH3, Bcl-2 homology 3 domain; BIM, Bcl-2-like protein 11; EP300, E1A-asosciated protein p300; ETV1, Ets variant gene 1; KIT, proto-oncogene tyrosine-protein kinase; MET, MET proto-oncogene receptor tyrosine kinase; mTOR, mammalian target of rapamycin complex 1; p21, cyclin dependent kinase inhibitor 1A; PLCG1, phospholipase c gamma 1; PSAP, prosaposin; p53, tumor protein p53; PTEN, phosphatase and tensin homolog; RB1, RB transcriptional corepressor 1; RHO, ROCK Rho-associated protein kinase; SLU7, pre-mRNA splicing factor SLU7; SMAD2, mothers against dpp homolog 2; TRCP2, F-box and WD repeat domain containing 11; TGF, transforming growth factor; WEE1, Wee1A kinase; Wnt, wingless-type mmtv integration site family.
Biomedicines 09 00463 g002
Table 1. Different tumor suppressing miRNAs downregulated and their targets in human osteosarcoma (OS) (data recovered by [61]).
Table 1. Different tumor suppressing miRNAs downregulated and their targets in human osteosarcoma (OS) (data recovered by [61]).
TargetmiRNAs
Aurora-BLet-7
ESR1miR-1
Bcl-2miR-15a, miR-34a, miR-125b, miR-143, miR-190b, miR-326, miR-449a
IGF systemmiR-16, miR-26a, miR-133a, miR-150, miR-497
MED27miR-18a
TAK1miR-20a
VEGFmiR20b, miR-29b, miR-205, miR-410
HMG systemmiR-22, miR-106a-5p, miR-142-3p, miR-505
SATB1miR-23a, miR-376a-3p
LPAATβmiR-24
SOX4, SOX9 or SOX2miR-25, miR-25-3p, miR-132, miR-188, miR-212, miR-32 (SOX9), miR-336 (SOX2)
PFKFB3miR-26
PTENmiR-30a
HIF1miR-33b, miR-20b, miR186
MARCKSmiR-34c-3p
EZRINmiR-96, miR-144, miR-150
TNFAIPmiR-15a, miR-99a,
PI3K-AKTmiR-100, miR-497
mTORmiR-101
Β-cateninmiR-107, miR-184
TGF βmiR-124, miR-153, miR-422a
TGF-αmiR-205, miR-376a, miR-376c, miR-422a,
ANXA2miR-206
ROR2miR-208b
ROCK1miR-129-5p, miR-139, miR-144, miR-145, miR-148a, miR-150, miR-198, miR-214-5p
CDK14miR-216a
CDK6miR-377, miR-494, miR-3928, miR-494
PDK1miR-379
LRH-1miR-381, miR-451
ELF2miR-409-3p
ZEB1miR-126, miR-130a, miR-141, miR-200b, miR-429, miR-643
SETD8miR-127-3p
Sirt1miR-133b, miR-138, miR-204
c-MycmiR-135b, miR-449c
MTDHmiR-136
FXYD6miR-137
HDAC4miR-140
GLI2miR-141-3p, miR-202
FASNmiR-142-3p
FOSL2miR-143-3p
E2F1miR-320
E2F3miR-152, miR-874
Wnt systemmiR-154, miR-184, miR-217
TIAM1miR-182
LRP6miR-183
ZERB2miR-187
TCF7miR-192
CDH2miR-194
NKD1miR-195-5p
VANGL-2miR-199-3p
PMP22miR-200bc/429
RAB22AmiR-203
ANXA2miR-206
ROR2miR-208b
Hsp90B1miR-223
Ect-2miR-223
Rac-1miR-224
ADAM9miR-302a
LDHAmiR-323a-3p
Rab10miR-329
Bmi-1 (or BMI1)miR-330-3p, miR-452
SurvivinmiR-335
AEG-1miR-342-3p, miR-448, miR-506
KRASmiR-365
FOXM1miR-370
FOXP4miR-491-5p
SATB1miR-376a-3p
LRH-1miR-451
c-MetmiR-454, miR-613
PKCmiR-486
HMGA2miR-490-3p
HMGB1miR-505
HMGN5miR-495
TSPAN1miR-491-3p
PAK6miR-492
ARL2miR-497-5p
FGF2miR-503
AKT-1 (or Akt)miR-520a-3p, miR-564
MMP8miR-539
ITGAVmiR-548c-3p
KLF5miR-590-5p
PDGFBmiR-598
YAP-1miR-625
PIM1miR-638
CYC1miR-661
RAB23miR-665
BCL9miR-1301
ERBB3miR-3928
Abbreviations: ESR1 (Estrogen Receptor 1), Bcl-2 (B Cell Lymphoma), IGF (Insulin Growth Factor), MED27 (Mediator Complex subunit 27), TAK1 (Mitogen-activated protein 3 kinase 7), VEGF (Vascular Endothelial Growth Factor), HMG (High Mobility Group), SATB1 (Special AT-rich sequence-binding protein 1), LPAATβ (Lysophosphatidic acid acyltransferase β), SOX (Sex Determining region Y-box), PFKFB3 (6-phosphofructo-2-kinase/fructuose-2,6-biphosphatase-3), PTEN (Phosphatase and Tensin homolog), HIF1 (Hypoxia-inducible transcription factor-1), MARCKS (Myristoylated alanine-rich protein kinase C substrate), TNFAIP (Tumor Necrosis Factor-α-induced protein), PI3K-AKT (Phosphatidil Inositol 3-Kinase), TGF (Transforming Growth Factor), ANXA2 (Annexin 2), ROR2 (Receptor Tyrosine kinase-like Orphan Receptor 2), ROCK1 (Rho associated Coiled-Coil containing protein Kinase 1), CDK (Cyclin-dependent kinase), PDK (Pyruvate Dehydrogenase Kinase), LRH-1 (Liver Receptor Homolog-1), ELF2 (E74-Like Factor 2), ZEB1 (Zinc Finger E-box Homeobox 1), Sirt1 (Sirtuin 1), MTDH (Metadherin), FXYD6 (FXYD Domain containing Ion Transport Regulator 6), HDAC4 (Histone Deacetylase 4), FASN (Fatty Acid Synthase), FOSL2 (FOS-Like antigen 2), TIAM1 (T-cell Lymphoma Invasion and Metastasis-inducing protein 1), LRP6 (Lipoprotein Receptor-related Protein 6), ZERB2 (Zinc finger E-box Binding Homeobox 2), TCF7 (T cell-specific transcription factor 7), CDH2 (Cadherin-2), NKD1 (Naked Cuticle Homolog 1), VANGL-2 (Van Gogh-Like 2), PMP22 (Peripheral Myelin Protein 22), RAB22A (Ras-related protein Rab-22A), Hsp90 (Heat Shock protein 90), Ect-2 (Epithelial Cell Transforming 2), LDHA (Lactate Dehydrogenase A), AEG-1 (Astrocyte-Elevated Gene 1), KRAS (Kirsten Rat Sarcoma viral oncogene), FOX (Forkhead Box Protein), PKC (Protein Kinase C), TSPAN1 (Tetraspanin 1), PAK6 (p-21 Activated Kinase 6), ARL2 (ADP Ribosylation factor-Like protein 2), FGF2 (Fibroblast Growth Factor 2), MMP (Matrix Metalloproteinase), ITGAV (Integrin Alpha V), KLF5 (Kruppel-Like Factor 5), PDGFB (Plateled Derived Growth Factor B), YAP-1 (Yes-associated protein 1), PIM1 (Moloney murine Leukemia virus 1), CYC1 (Cyclin 1), RAB23 (Ras-related protein Rab-23), BCL9 (B-cell CLL/lymphoma 9), ERBB3 (erb-b2 receptor tyrosine kinase 3).
Table 2. Different onco-miRNAs upregulated and their targets in human OS (data recovered by [61]).
Table 2. Different onco-miRNAs upregulated and their targets in human OS (data recovered by [61]).
TargetmiRNAs
GCIPmiR-9
KLF4miR-10b
PTENmiR-17, miR-21, miR-23a, miR-92a, miR-93, miR-196a, miR-214, miR-221, miR-1908
BRCC2miR-17-5p, miR-603
SOCS6miR-19
EGR2miR-20a
ING5miR-27-3p
TET1miR-27a-3p
TGF-βmiR-29
RASSF1AmiR-81a
RECKmiR-92b
CDKN1miR95-3p
VNN2miR-106a
PI3KmiR-106b
TPM1miR-107
PPARγmiR-130b
CDC14AmiR-131a
ROCK1miR-138
BMP9miR-149
HBP1miR-155
RIPK1miR-155-5p
CFIm25miR-181a
Chk2miR-191
p21miR-93, miR-95-3p
p27miR-199a-5p
BRD7miR-300
DAB2IPmiR-367
FOXO1 and FOXO4miR-374a, miR-660, miR-664 (FOXO4)
KLF9miR-378
MTSS1miR-411
BimmiR-488
TP53INP1miR-504
VANGL-2miR-542-3p, miR-542-5p
MAP3K9miR-1247
AGTR1miR-1248
SRSF3miR-1908-5p
Abbreviations: GCIP (Glutamate receptor-interacting protein), KLF4 (Kruppel-Like Factor 4), PTEN (Phosphatase and Tensin homolog), BRCC2 (Breast Cancer Cell 2), SOCS6 (Suppressor of Cytokine Signaling 6), EGR2 (Early Growth Response 2), ING5 (Inhibitor of Growth Family member 5), TET1 (Ten Eleven Translocation 1), TGF-β (Transforming Growth Factor β), RASSF1A (Ras Association domain Family 1 isoform A), RECK (Reversion inducing Cysteine Rich protein with Kazal motifs), CDKN1 (Cyclin Dependent Kinase inhibitor 1), VNN2 (Vanine 2), PI3K (Phosphatidil Inositol 3 Kinase), TPM1 (Tropomyosin 1), PPARγ (Peroxisome Proliferator-Activated Receptor γ), CDC14A (Dual specificity protein phosphatase 14A), ROCK1 (Rho associated Coiled-Coil containing protein Kinase 1), BMP9 (Bone Morphogenetic Protein 9), HBP1 (HMG-Box Transcription Factor 1), RIPK1 (Receptor-Interacting serine/threonine-Protein Kinase 1), CFIm25 (Component of the Cleavage Factor Im25), Chk2 (Checkpoint Kinase 2), BRD7 (Bromodomain Containing 7), DAB2IP (Disable homolog 2-Interacting Protein), FOXO (Forkhead box Protein O), KLF9 (Krueppel-Like Factor 9), MTSS1 (MTSS I-BAR Domain Containing 1), Bim (Bcl-2-Like Protein 11), TP53INP1 (Tumor Protein p53-Inducible Nuclear Protein 1), VANGL-2 (VANGL Planar Cell Polarity Protein 2), MAP3K9 (Mitogen-Activated Protein Kinase Kinase Kinase 9), AGTR1 (Angiotensin II Receptor Type 1), SRSF3 (Serine and Arginine Rich Splicing Factor 3).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Llobat, L.; Gourbault, O. Role of MicroRNAs in Human Osteosarcoma: Future Perspectives. Biomedicines 2021, 9, 463. https://doi.org/10.3390/biomedicines9050463

AMA Style

Llobat L, Gourbault O. Role of MicroRNAs in Human Osteosarcoma: Future Perspectives. Biomedicines. 2021; 9(5):463. https://doi.org/10.3390/biomedicines9050463

Chicago/Turabian Style

Llobat, Lola, and Olivia Gourbault. 2021. "Role of MicroRNAs in Human Osteosarcoma: Future Perspectives" Biomedicines 9, no. 5: 463. https://doi.org/10.3390/biomedicines9050463

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