Open access peer-reviewed chapter

Role of miRNA in Glioma Pathogenesis, Diagnosis, and Therapeutic Outcomes

Written By

Manendra Singh Tomar and Ashutosh Shrivastava

Submitted: 06 June 2023 Reviewed: 06 June 2023 Published: 29 June 2023

DOI: 10.5772/intechopen.1001998

From the Edited Volume

Molecular Biology and Treatment Strategies for Gliomas

Terry Lichtor

Chapter metrics overview

58 Chapter Downloads

View Full Metrics

Abstract

Glioma is the most aggressive tumor of glial cells in the brain and spinal cord. It comprises 30% of all brain tumors. Even in the presence of current multimodal therapeutic regimens, the life expectancy of more than 2 years is very rare. MicroRNAs (miRNAs) are short, non-coding RNAs produced naturally in the body and control gene expression. Many evidence-based hypotheses show that miRNA expression is aberrantly influenced in glioma due to amplification or deletion of miRNA genes, inappropriate transcriptional regulation of miRNAs, dysregulated epigenetic alterations, or faults in the miRNA biogenesis machinery. In some circumstances, miRNAs promote tumorigenesis, whereas under other circumstances, they can act as tumor suppressors in glioma. In gliomas, miRNA is involved in cell proliferation signaling, evasion of growth suppressors, resistance to cell death, tumor cell infiltration, metastasis, and angiogenesis. More and more research is pointing to miRNAs as prospective biomarkers for glioma diagnosis, prognosis, and treatment targets or tools; however, these claims have yet to be validated. Here, we discuss the role of microRNAs (miRNAs) as tumor suppressors and oncogenes in the growth of glioma.

Keywords

  • MiRNA
  • glioma
  • carcinogenesis
  • diagnostic biomarkers
  • therapeutics

1. Introduction

Gliomas are the primary brain neoplasm of the central nervous system (CNS) hypothesized to develop from neuroglial stem or progenitor cells. They have histologically been categorized as astrocytic, oligodendroglial, or ependymal tumors based on their histological appearance and accordingly given WHO categories I–IV, which indicate varying degrees of aggressiveness [1]. Due to the advent of new prognostic markers of glioma, WHO updated their classification based on new molecular markers in 2016 [2, 3]. Glioblastoma multiforme (GBM) has a high degree of lethality and low median survival of ~15 months after the initial diagnosis and the five-year survival is less than 10% [4]. The centerpiece of GBM treatment is surgery, followed by radiation and adjuvant chemotherapy. The blood-brain barrier (BBB) severely restricts therapeutic agent delivery to the CNS. Therefore, the accessibility of the drug to the tumor sites is the main obstacle for the development of new therapeutics for GBM [5]. Temozolomide (TMZ) is a first-choice alkylating agent inducted as a standard therapy for glioblastoma multiforme (GBM) and astrocytoma. TMZ has limited efficacy due to the development of intrinsic and extrinsic drug resistance in glioma [6].

Growing shreds of evidence suggest that microRNA (miRNA) has a role in the onset and progression of gliomas [7]. miRNAs were first identified in the nematode Caenorhabditis elegans but have now been detected in almost every eukaryotic organism. MicroRNA is postulated to influence around 30% of human genes that code for proteins and make up 1–5% of the human genome [8]. These are short, evolutionarily conserved, single-stranded, non-coding RNA molecules that bind the target mRNA and inhibit protein generation [8]. Thus, it is not surprising that numerous miRNAs with a function in controlling apoptosis have also been connected to the emergence and development of cancer and other neoplastic diseases. Half of all the miRNAs found in genomic regions are often altered or amplified in malignant tumors [9]. Moreover, miRNAs have been linked to many aspects of carcinogenesis, including their invasiveness, DNA repair, and acquired resistance. Due to their numerous functions, miRNAs could replace other classes of molecules as primary targets of therapy for glioma [10]. Different miRNAs have opposing roles as tumor suppressors and oncogenes. Moreover, signaling that promotes chemotherapy resistance and tumor promotion in glioma is also triggered by the alteration or overexpression of oncogenic miRNAs and the downregulation of tumor-suppressing miRNAs [11]. In addition to providing a bad prognosis for glioma, recent research found that cytosine methylation of mature miRNA decreases its tumor-suppressive ability [12].

Continued research on miRNA is critical to understand its functional role in early diagnosis and treatment of GBM. This book chapter provides an overview of the pivotal role of miRNAs in glioma development, with special emphasis on their function as a diagnostic biomarker and their therapeutic potential.

Advertisement

2. MicroRNA biogenesis

The biogenesis of miRNA is classified into canonical and non-canonical pathways [13]. Most of the miRNAs are processed using the well-established canonical biogenesis mechanism. The microprocessor complex, comprised of the RNA-binding protein DiGeorge Syndrome Critical Region 8 (DGCR8) and the ribonuclease III enzyme Drosha, converts pri-miRNAs generated from their genes into pre-miRNAs [14]. DGCR8 recognizes N [6] methyl adenosine mark, a type of post-transcriptional modification that promotes the initiation of miRNA biogenesis [15]. RNase III enzyme DROSHA recognizes and cleaves miRNA hairpins, resulting in the release of pre-miRNAs, i.e., miRNAs with a length of 60–80 nucleotides [16]. The GTP-binding nuclear protein Ran (RanGTP)/exportin-5 (XPO5) complex transports pre-miRNAs from the nucleus to the cytoplasm, where Dicer processes them into mature miRNAs with functional and regulatory capabilities (Figure 1) [17]. Generally, mature miRNA has one guide strand and the other passenger strand. Next, an argonaute protein takes the miRNA duplex and forms a structure called the miRNA-Induced Silencing Complex (RISC) [18]. The RISC is a multiprotein complex that contains a single strand of miRNA. RISC acts as a template for the recognition of complementary mRNA based on the miRNA. Upon recognizing a matching strand, it triggers RNase and cleaves the RNA [19].

Figure 1.

Canonical and non-canonical pathway of miRNA biogenesis.

Non-canonical pathways begin with shRNA being cleaved by the microprocessor complex and then exported to the cytoplasm through the Exportin5/RanGTP complex. They undergo further processing that requires Argonaute 2 (AGO2) but does not need Dicer [20]. AGO2 is an RNA-binding protein that plays a crucial role in RNA-silencing processes. It regulates chromatin structure, transcriptional gene control, and RNA splicing, and plays a critical role in the development and function of microRNAs [21]. Two different types of miRNAs are processed through the non-canonical pathway. One of them, called mirtrons, is generated from an mRNA intron during the splicing process and the other is pre-miRNA that is capped with 7-methylguanosine (m7G) [2022]. A functioning miRISC complex is the endpoint of all possible routes that bind to mRNA and leads to translational inhibition (Figure 1) [23].

Advertisement

3. MicroRNA in the pathogenesis of glioma

Over the last several decades, researchers have worked hard to catalog miRNAs with aberrant expression patterns in glioma and choose the most promising ones for further evaluation as potential therapeutic targets. Multiple studies have shown a wide variety of miRNAs that play important roles in gliomas [24]. Ten signature miRNAs were described through TCGA analysis of mRNA expression data that may be crucial to distinguish between low- and high-risk GBM patients. Three of which were tumor-suppressing (miRNA-20a, miRNA-106a, and miRNA-17-5p), and seven of which were oncogenic (miRNA-31, miRNA-222, miRNA-148a, miRNA-221, miRNA-146b, miRNA-200b, and miRNA-193a) [25]. A significant proportion of miRNAs were observed to be overexpressed in glioma compared to normal brain tissue, 95 have relatively lower expression, and inconsistent findings were detected for 17 miRNAs [26].

3.1 MicroRNAs in glioma cell proliferation, migration, and invasion

Tumor-suppressing miRNAs are progressively linked to the advancement of antitumor treatment, via remodeling gene networks that are dysregulated in malignant tumors. The role of miRNA-21 is most extensively studied in glioma and is consistently upregulated. It inhibits cell death and boosts tumor invasion. It performs a role in chemo- and radiation resistance [27]. Similarly, miRNA-138 is one of the most significant miRNAs in GBM patients and correlates inversely with CD44 expression. Also, cell cycle arrest happens in GBM cells after overexpression or administration of miRNA-138, which negatively affects the expression of CD44 by directly binding to its 3′ UTR [28]. Highly expressed and exceptionally abundant miRNAs are found in glioma associated-human mesenchymal stem cells (GA-hMSCs)-derived exosomes, and one of them, miR-1587, has been shown to dramatically boost Glioma Stem Cell proliferation and clonogenicity. Exosomes generated from GA-hMSCs carry miR-1587 and cause the downregulation of tumor suppressor nuclear receptor corepressor NCOR1 resulting in the increased aggressiveness of GBM [29]. Additionally, miRNA-3620-5p enhances tumor cell proliferation and promotes stemness in glioma [30]. A Phosphatase and Tensin homolog (PTEN) regulating miRNA-26a is amplified in high-grade glioma and promotes tumor generation [31].

Other classes of miRNAs are also reported whose upregulation suppresses tumor cell proliferation such as miRNA-1825. This miRNA was found to be decreased in GBM patients, whereas its upregulation suppresses tumor progression by targeting CDK-14 through the Wnt/β-catenin signaling pathway [32]. According to another study, miRNA-10a regulates Wnt/β-catenin signaling and induces tumor generation via targeting myotubularin-related protein 3 [33]. In glioma tissues, decreased levels of miRNA-382-5p significantly increased the proliferation, invasion, and migration of tumor cells. Conversely, the transfer of mimics of miRNA-382-5p significantly decreases neoplasm formation as well as epithelial-to-mesenchymal transition (EMT) [34]. It has been established that the EMT plays a crucial role in the spread of tumors. The miRNA-16 suppresses EMT mainly through the downregulation of p-FAK and p-Akt expression and NF-kB and slug transcriptional activity [35].

During tumorigenesis, many proteolytic enzymes, including matrix metalloproteinases (MMPs), are overexpressed. Their job is to dissolve and degrade the matrix proteins of the surrounding normal brain tissue, which act as a barrier to migration and invasion of tumor cells [36]. Compared to healthy controls, glioma patients express more MMP-9. Further studies revealed that the expression status of MMP-9 and miRNA-34a was inversely correlated. Overexpression of miRNA-34a suppressed cell proliferation and migration in human glioma cells by MMP-9 [37]. Moreover, in GBM, miRNA-25 is elevated, and its expression levels correlate strongly with disease progression. miRNA-25 silencing significantly reduces tumor cell movement and invasiveness, by increasing the expression of cell adhesion molecule 2 (CADM2). This establishes CADM2 as a suitable target for miRNA-25 since CADM2 is dramatically downregulated in glioma [38]. On the other hand, miR-146b-5p, a tumor-suppressing miRNA, has been shown to have a negative correlation with its target gene MMP16. Glioma cells exhibit considerably lower amounts of miR-146b-5p, while simultaneously displaying higher levels of MMP16. The excessive production of miR-146b-5p also facilitates the disintegration of mRNA associated with MMP16 as well as restored miRNA action against glioma cell migration and invasion [39].

Cancer patients also have abnormal TGF-β signaling, and this abnormal signaling likely plays a role in the development and progression of several malignancies, including glioma [40]. A correlation study showed a positive relationship between TGF-β concentration and miRNA-132 mRNA levels. Additionally, it is reported that miRNA-132 specifically targets SMAD7 3’-UTR, resulting in downregulation of SMAD7 expression and modification of the TGF-β signaling pathway. Patients with glioma have an inverse relationship between miRNA-132 and SMAD7 [41]. Degradation or lower level of SMAD7 promotes autophagy-induced EMT and chemoresistance in glioma [42]. Evidence from these investigations shows that aberrant miRNA expression has a major role in tumor cell growth, migration, and invasion. All these findings strengthen the case for miRNAs as a potential therapeutic target in glioma.

3.2 MicroRNAs and angiogenesis network in glioma

The development of GBM is reliant on the establishment of new blood vessels since gliomas are highly vascularized tumors [43]. In response to various stimuli, vascular endothelial cells (ECs) proliferate, migrate, and differentiate to form new blood vessels, a process known as angiogenesis [44]. More intriguing is the discovery of miRNAs regulating angiogenesis via necessary biochemical pathways and its activation and/or inhibition. These miRNAs are known as “angiomiRs,” and they play important functions in regulating the vascular network characteristics of gliomas [30].

Primary tumor endothelial cells of glioma have higher miR-296 levels than normal brain cells. Growth factor-induced miR-296 directly targets the hepatocyte growth factor-regulated tyrosine kinase substrate (HGS) mRNA, decreasing HGS levels and reducing HGS-mediated degradation of the growth factor receptors VEGFR2 and PDGFR-β. This activates angiogenesis in glioma cells [45]. Also, dysregulation of miRNA-24 in glioma cells promotes microvascular proliferation of endothelial cells as well as enhances the expression of molecules related to angiogenesis like MMP-2, 9, VEGF, and TGF-β [46]. Additionally, miRNA-101 is downregulated in GBM as a result of that Enhancer of zeste homolog 2 (EZH2)-induced proliferation, migration, and angiogenesis. Inhibition of EZH2 in glioma reduced angiogenesis and tumor growth [47].

Several miRNAs have been identified to play a crucial role in angiogenesis, and the transfer of RNAs from GBM cells to brain endothelial cells through extracellular vesicles induces angiogenesis. miRNA-148a and miRNA-9-5p both are linked with glioma angiogenesis and poor patient survival. When miRNA-9 is supplied to human umbilical ECs (HUVEC) through GBM-derived EVs, the number and length of tubules that develop as a consequence are inextricably linked with miRNA-9 expression in HUVECs [48, 49]. Transcriptional study identified RGS5, SOX7, and ABCB1 as a miRNA-9 target. Out of these, RGS5 and SOX7 are crucial for angiogenesis and cellular proliferation [48, 50, 51]. In experimental models of GBM, suppressing miRNA-148a normalizes the abnormal tumor angiogenesis [52].

Similarly, GSCs-derived miRNA-26a promotes proliferation, migration, and angiogenesis through inhibition of PTEN [53]. Vascular endothelial growth factor (VEGF) and VEGF receptor transcriptional activity may be controlled by the transcription factor Myc-associated zinc finger protein (MAZ). MAZ expression is regulated through miRNA-125; consequently, miR-125b overexpression prevents tumor angiogenesis [54, 55]. Several other miRNAs are also identified such as miRNA-124-3p, miRNA-15b, and miRNA-152 that play important roles in glioma cell invasion and angiogenesis. Neuropilin-1 (NRP-1) promotes GBM cell development and growth that suppress miRNA-124-3p expression [56], while miRNA-15b and miRNA-152 inhibit glioma cell migration and angiogenesis through NRP-2 and MMP-3, respectively [57].

3.3 MicroRNAs in the generation of glioma stem cells

When compared to normal brain samples, GBM contains dysregulated levels of many miRNAs that play a crucial role in the generation of stem cell-like properties [26]. Loss of function assay of miRNA-663a induces proliferation, migration, invasion as well as stem cell-like properties in glioma. Gain of function assay shows overexpression of miRNA-663a represses cancer stem cell-like properties via inhibiting lysine demethylase 2A-mediated TGF-β/SMAD signaling pathway [58]. MiRNA-7 controls master regulators like the HoxD family of proteins as well as particular neuron activities including synaptic transmission. In line with its function in differentiation and proliferation, it has been identified as one of the miRNAs comprising the “miRNA signature” in neural stem and neural cancer stem cells [59]. The miRNA-9 is much more expressed in GBM CSF, GSCs, and GSC-derived EVs, which contributes to the aggressiveness of the disease; however, miRNA-9 inhibition had the opposite anticancer impact [60].

A microarray-based screening of 455 high-grade glioma patients identified differences in the level of miRNA-10a, miRNA-10b, and miRNA-139 that are crucial for the growth and differentiation of GSCs [61]. Enhanced differentiation of neurons was seen in murine astrocytic neural stem cells, murine oligodendroglioma-derived S100-v-erbB+ stem cells, human GBM CD133+ CSCs, and GBM cell lines after transfection with miRNA-124a or miRNA-137. In addition, G0/G1 cell cycle arrest and reduced expression of cell cycle components CDK6 and phosphorylated Rb were seen after co-transfection of miRNA-124a and miRNA-137 in GBM [62]. The miRNA-302-367 inhibits clonal development and SHH/GLI1/NANONG network regulation in GBM cells by targeting CXC chemokine receptor type 4 (CXCR4) and its ligand, stromal-derived factor 1 (SDF1). These findings suggest that miRNA-302-367 regulated a key pathway essential for the regeneration of neuron and stem cell phenotype [62]. Downregulation of miRNA-29b and miRNA-125a in CD133+ GSCs was also documented [63]. Similarly, like miRNA-125a, miRNA-125b plays a critical function in controlling the expansion of GBM cells and the activity of CSCs. A reduction in miRNA-125b in GBM cells was seen after all-trans-retinoic acid treatment [64].

Several other studies also identified miRNA dysregulation in GSCs compared to GBMs. Recent studies found miRNA-128a-3p, 34-5p, and 181a-3p to be downregulated, whereas miRNA-17-5p and miRNA-221-3p were increased in stem-like cells [65]. Also, miRNA-145 was typically under-expressed in GCSs, and its upregulation enhances chemosensitivity and cellular apoptosis [66]. MiRNA-27a-3p, miRNA-22-3p, and miRNA-221-3p were delivered to GSCs by EVs from monocyte-derived macrophages, and by concurrently targeting CHD7. These miRNAs induced proneural-to-mesenchymal transition in GSCs [67]. The miRNA expression profile reveals MiR-9, miR-9(*), miR-17, and miR-106b to be significantly abundant in CD133(+) cells, which also include populations of GSCs [68].

3.4 MicroRNAs in drug resistance in glioma

Several mechanisms of drug resistance include dysfunctional DNA repair, overexpression of drug efflux transporters, apoptosis inhibition, and increased epithelial-to-mesenchymal transition. The expression levels of MDR transporters are significantly impacted by miRNAs, which play a crucial role in controlling glioblastoma MDR [69]. Among the ABC transporter family, ABCG2 is particularly prominent in glioblastoma [70]. Downregulation of miRNA-328 reduces ABCG2 expression and chemoresistance in glioblastoma, suggesting that miRNA-328 plays a critical role in ABCG2 expression [71]. Therefore, miR-328 therapy combined with radiation or chemotherapy may be a useful approach for treating GBM [72]. Likewise, miRNA-1268a is downregulated after the TMZ treatment as a result of its target membrane transporter ABCC1/MRP1 being upregulated. Contrarily, overexpression of miRNA-1238a inhibits protein translation of ABCC1 and restores sensitivity to Temozolomide (TMZ) [73]. Moreover, by repressing miR-128-3p’s effects on MRP1, RUNX1 makes GBM resistant to TMZ. Overcoming TMZ resistance in GBM may be possible via targeting the miR-128-3p/RUNX1/MRP1 axis [74].

Reversal of MDR in glioblastoma cells is also described after overexpression of miR-9 levels, which is associated with the suppression of ABC transporters such as MDR1, ABCC3, and ABCC6 [75]. In GBM, miRNA-381 was shown to be overexpressed; blocking miRNA-381 or driving neurofilament light polypeptide (NEFL) expression and greatly sensitizing GBM cells to the chemotherapeutic drug TMZ. Multiple multidrug resistance proteins (ABCG2, ABCC3, and ABCC5) and stemness factors (ALDH1, CD44, CKIT, KLF4, Nanog, Nestin, and SOX2) are the possible target of miRNA-381 by which these cells are sensitized to TMZ [76]. The ABCB1 is another drug efflux transporter that is overexpressed in resistant GBM cells and the study identified that expression of the transporter is controlled through miRNA-4539 and miRNA-4261 [77]. The aforementioned research emphasizes the importance of miRNA in ABC transporter-mediated drug efflux and suggests it as a viable therapeutic target for reestablishing chemosensitivity in gliomas.

Temozolomide is a first-choice alkylating agent inducted as a standard therapy for GBM and astrocytoma [78]. TMZ has limited efficacy due to the development of intrinsic and extrinsic drug resistance [79]. TMZ is an alkylating agent that generates methyldiazonium cation transfers their methyl group on N7 and O6 sites of guanine and the N3 on adenine [80]. The TMZ adduct imparts mutation in DNA fixed via O6-methylguanine-DNA methyltransferase (MGMT), Mismatch Repair (MMR), and Base Excision Repair (BER). MMR generates DNA double-strand breaks (DSBs) in TMZ-sensitive cells and triggers PCD, whereas overexpression of MGMT and other repair proteins remove TMZ adducts and generate TMZ-resistant glioma cells [11].

DNA mismatch repair (MMR) is a mechanism that detects and repairs DNA replication and recombination errors, including insertions, deletions, and misincorporations of nucleotides [81]. Compared to other cell types, radioresistant glioma cells had considerably high levels of DNA2 and low levels of miR-3059-3p. It has been identified that miR-3059-3p regulates radioresistance by targeting DNA2 and controlling homologous recombination pathways through Rad51 & 52. MiR-3059-3p-mediated downregulation of DNA2 inhibited the HR pathway, making GBM cells more radiosensitive [82]. Glioma cell proliferation is inhibited and senescence-related genes p53, Cdkn1a, and Cdkn2c are upregulated after delivery of miR-34a. Increased levels of miR-34a also reduce telomerase activity, shorten telomeres, and cause DNA damage. Forced overexpression of SIRT1 can counteract these pro-senescent effects [83].

Patients with GBM exhibited downregulation of miRNA-198 [84]. Reduced levels of these miRNAs are associated with a worse outcome for patients. Increased chemosensitivity to TMZ is also associated with elevated expression of miRNA-198, as shown by in vitro and in vivo investigations. This was achieved by miRNA-198 inhibiting protein translation specifically of MGMT [85]. Likewise, the miRNA-370-3p, which is suppressed in GBM, when transfected into TMZ-resistant GBM cells, makes them more sensitive to the anticancer medication by reducing their ability to fix their genetic lesions. Studies suggest that MGMT is a direct target of miRNA-370-3p and this miRNA plays a crucial role in the restoration of chemoresistance in GBM [86].

Additionally, multiple studies have shown the role of miRNAs in GBM tumorigenesis and onset of chemoresistance via activation of base excision repair proteins such as PARP, XRCC, Rad51, and others [87]. miRNA-151a depletion led to the development of TMZ resistance. And according to another study, enhanced miRNA-151a production sensitizes TMZ-resistant GBM cells by decreasing XRCC4-mediated DNA repair [88].

Advertisement

4. MicroRNA as glioma diagnostic biomarker

As we discussed earlier, miRNAs act as either oncogenes or tumor suppressors in glioma. Changes in the expression of these molecules have been linked to several cancer types, making them a promising molecular tool for non-invasive cancer detection and prognosis [89]. Hereby, we list some of the miRNAs that could be of diagnostic and prognostic significance (Table 1).

S.No.Name of miRNAsSample TypeMarker TypeReference
1.miRNA-21.Tissue, Plasma, EVs, CSF, plasma, and serum.Diagnostic and Prognostic.[90, 91, 92, 93]
2.miRNA-128, miRNA-342-3p.PlasmaDiagnostic and Prognostic.[93]
3.miRNA-221Tissue, CSF, Plasma, and serum.Diagnostic[90, 94, 95]
4.miRNA-451EVsDiagnostic[96]
5.miRNA-182Tissue and Plasma.Diagnostic[97, 98]
6.miRNA-27aTissue, EVs, and CSF.Prognostic[99, 100]
7.miRNA-21-5p, miRNA-218-5p, miRNA-193b-3p, miRNA-331-3p, miRNA-374a-5p, miRNA-548c-3p, miRNA-520f-3p, miRNA-27b-3p, miRNA-30b-3p.CSFDiagnostic, Prognostic.[101]
8.miRNA-125, miRNA-222.CSF, Blood.Diagnostic[102]
9.miRNA-10bTissue, CSF, Serum.Diagnostic, Prognostic.[92, 103]
10.miRNA-183TissuePrognostic[104]

Table 1.

MiRNAs as a potential diagnostic and prognostic biomarker in glioma.

Advertisement

5. Therapeutic potential of miRNAs in glioma

Overall, the findings described in this chapter show that miRNAs are implicated in many pathways that worsen glioma patients’ prognoses by promoting tumor growth, invasion, and resistance to therapy. What needs to be examined are the translational implications of these results and strategies to utilize this information in clinical practice. As we are aware, cancer is a complex disease that develops and spreads via the involvement of a variety of oncogenes and signaling mechanisms. The capacity of miRNA to concurrently target several genes involved in the same biochemical process is one of the numerous potential advantages of miRNA as a therapeutic agent. The two therapeutic focuses of miRNA-based treatment are miRNA replacement or antisense therapy and modulation of miRNA biogenesis [105]. DNA methylation and histone modification are two examples of epigenetic modulation that regulate certain miRNA biogenesis in cancer cells. Tumor-suppressing miRNAs can be activated with chromatin-modifying medications to control target oncogenes, which could pave the way to new cancer treatments in the near future [106]. A further advantage of miRNA mimics or replacement therapy is that they are predicted to target the same group of mRNAs that are controlled by the reduced original miRNA since they share the same sequence. Due to the designed imitation of miRNA mimics to replicate the behavior of their natural counterparts, the likelihood of experiencing any unintended off-target consequences is extremely low [107].

Scientists are actively investigating how to alter synthetic miRNAs to facilitate better translocation to host cells in vivo to increase the effectiveness of miRNAs in the field of cancer therapies. Several methods, including viral and non-viral alterations and chemical ones, are proposed to improve target delivery. For example, it is less probable for synthetic miRNAs to be degraded by nucleases if specific structural components, such as the 2’-OH of ribose or the phosphate backbone, are changed [108]. The cellular miRNAs’ processing capacity is often poor and they are vulnerable to nuclease degradation, which reduces their bioavailability.

Recent studies also employ polymer nanoparticles as delivery vehicles for microRNAs (miRNAs) in GBM. Commonly used polymers include Poly (lactic-co-glycolic acid) or PLGA and Polyethyleneimine (PEI). Increased TMZ chemosensitivity was seen in both in vitro and in vivo after the delivery of antimiR-21 and antimiR-10b into GBM cells using PLGA nanoparticles [109, 110, 111]. Protecting miRNAs against degradation in a physiological environment is greatly aided by lipid nanoparticle delivery methods. Lipid nanoparticles are advantageous for transporting miRNAs in the clinic because of this quality. Complexes are readily formed when positively charged lipids are combined with negatively charged miRNAs through electrostatic interactions. These complexes increase the absorption rates of miRNAs [5, 112]. It is anticipated that if the delivery barrier is broken down and gaining a deeper knowledge of the effect and longevity of gene silencing, miRNAs are predicted to become useful therapies in the clinic.

Advertisement

6. Conclusion

Genome-wide profiling shows that miRNA expression is linked to tumor subtype, tumor grade, and patient outcomes in different cancers including glioma. Hence, miRNAs can be used as diagnostic, prognostic, and therapeutic biomarkers. Since it has been shown that miRNA deletion or overexpression is associated with glioma, researchers all over the globe have been trying to determine what functions miRNA play in cancer and what causes their expression to be dysregulated. MicroRNAs are hypothesized to behave as oncogenes or tumor suppressors in glioma by modifying particular targets. Numerous therapeutically promising miRNA antagonists and miRNA analogs are now in clinical trials, but further work is required to verify them as diagnostic and prognostic biomarkers in a large cohort of patient samples.

Advertisement

Acknowledgments

Manendra Singh Tomar is the recipient of a Junior research fellowship from the University Grants Commission, Government of India.

Advertisement

Declaration of interest

Authors declare no conflict of interest.

References

  1. 1. Weller M, Wick W, Aldape K, Brada M, Berger M, Pfister SM, et al. Glioma. Nature Reviews. Disease Primers. 2015;1:15017. DOI: 10.1038/nrdp.2015.17
  2. 2. Tomar MS, Shrivastava A. TERT promoter mutations correlate with IDHs, MGMT and EGFR in glioblastoma Multiforme. Neurology India. 2021;69(1):135-136. DOI: 10.4103/0028-3886.310071
  3. 3. Brito C, Azevedo A, Esteves S, Marques AR, Martins C, Costa I, et al. Clinical insights gained by refining the 2016 WHO classification of diffuse gliomas with: EGFR amplification, TERT mutations, PTEN deletion and MGMT methylation. BMC Cancer. 2019;19(1):968. DOI: 10.1186/s12885-019-6177-0
  4. 4. Hanif F, Muzaffar K, Perveen K, Malhi SM, Simjee Sh U. Glioblastoma Multiforme: A review of its epidemiology and pathogenesis through clinical presentation and treatment. Asian Pacific Journal of Cancer Prevention. 2017;18(1):3-9. DOI: 10.22034/apjcp.2017.18.1.3
  5. 5. Chen M, Medarova Z, Moore A. Role of microRNAs in glioblastoma. Oncotarget. 2021;12(17):1707-1723. DOI: 10.18632/oncotarget.28039
  6. 6. Lee SY. Temozolomide resistance in glioblastoma multiforme. Genes & Diseases. 2016;3(3):198-210. DOI: 10.1016/j.gendis.2016.04.007
  7. 7. Bartel DP. Metazoan MicroRNAs. Cell. 2018;173(1):20-51. DOI: 10.1016/j.cell.2018.03.006
  8. 8. Macfarlane LA, Murphy PR. MicroRNA: Biogenesis, function and role in cancer. Current Genomics. 2010;11(7):537-561. DOI: 10.2174/138920210793175895
  9. 9. Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, et al. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(9):2999-3004. DOI: 10.1073/pnas.0307323101
  10. 10. Banelli B, Forlani A, Allemanni G, Morabito A, Pistillo MP, Romani M. MicroRNA in glioblastoma: An overview. International Journal of Genomics. 2017;2017:7639084. DOI: 10.1155/2017/7639084
  11. 11. Tomar MS, Kumar A, Srivastava C, Shrivastava A. Elucidating the mechanisms of Temozolomide resistance in gliomas and the strategies to overcome the resistance. Biochimica Et Biophysica Acta. Reviews on Cancer. 2021;1876(2):188616. DOI: 10.1016/j.bbcan.2021.188616
  12. 12. Cheray M, Etcheverry A, Jacques C, Pacaud R, Bougras-Cartron G, Aubry M, et al. Cytosine methylation of mature microRNAs inhibits their functions and is associated with poor prognosis in glioblastoma multiforme. Molecular Cancer. 2020;19(1):36. DOI: 10.1186/s12943-020-01155-z
  13. 13. Miyoshi K, Miyoshi T, Siomi H. Many ways to generate microRNA-like small RNAs: Non-canonical pathways for microRNA production. Molecular Genetics and Genomics. 2010;284(2):95-103. DOI: 10.1007/s00438-010-0556-1
  14. 14. Denli AM, Tops BB, Plasterk RH, Ketting RF, Hannon GJ. Processing of primary microRNAs by the microprocessor complex. Nature. 2004;432(7014):231-235. DOI: 10.1038/nature03049
  15. 15. Alarcón CR, Lee H, Goodarzi H, Halberg N, Tavazoie SF. N6-methyladenosine marks primary microRNAs for processing. Nature. 2015;519(7544):482-485. DOI: 10.1038/nature14281
  16. 16. Lee D, Shin C. Emerging roles of DROSHA beyond primary microRNA processing. RNA Biology. 2018;15(2):186-193. DOI: 10.1080/15476286.2017.1405210
  17. 17. Wu K, He J, Pu W, Peng Y. The role of Exportin-5 in MicroRNA biogenesis and cancer. Genomics, Proteomics & Bioinformatics. 2018;16(2):120-126. DOI: 10.1016/j.gpb.2017.09.004
  18. 18. Medley JC, Panzade G, Zinovyeva AY. microRNA strand selection: Unwinding the rules. Wiley Interdisciplinary Reviews RNA. 2021;12(3):e1627. DOI: 10.1002/wrna.1627
  19. 19. Zhang Y. RNA-induced silencing complex (RISC). In: Dubitzky W, Wolkenhauer O, Cho K-H, Yokota H, editors. Encyclopedia of Systems Biology. New York, New York, NY: Springer; 2013. DOI: 10.1007/978-1-4419-9863-7_329
  20. 20. O'Brien J, Hayder H, Zayed Y, Peng C. Overview of MicroRNA biogenesis, mechanisms of actions, and circulation. Frontiers in Endocrinology (Lausanne). 2018;9:402. DOI: 10.3389/fendo.2018.00402
  21. 21. Tarallo R, Giurato G, Bruno G, Ravo M, Rizzo F, Salvati A, et al. The nuclear receptor ERβ engages AGO2 in regulation of gene transcription, RNA splicing and RISC loading. Genome Biology. 2017;18(1):189. DOI: 10.1186/s13059-017-1321-0
  22. 22. Ruby JG, Jan CH, Bartel DP. Intronic microRNA precursors that bypass Drosha processing. Nature. 2007;448(7149):83-86. DOI: 10.1038/nature05983
  23. 23. Lam JK, Chow MY, Zhang Y, Leung SW. siRNA versus miRNA as therapeutics for gene silencing. Molecular Therapy Nucleic Acids. 2015;4(9):e252. DOI: 10.1038/mtna.2015.23
  24. 24. Mafi A, Rahmati A, Babaei Aghdam Z, Salami R, Salami M, Vakili O, et al. Recent insights into the microRNA-dependent modulation of gliomas from pathogenesis to diagnosis and treatment. Cellular & Molecular Biology Letters. 2022;27(1):65. DOI: 10.1186/s11658-022-00354-4
  25. 25. Srinivasan S, Patric IR, Somasundaram K. A ten-microRNA expression signature predicts survival in glioblastoma. PLoS One. 2011;6(3):e17438. DOI: 10.1371/journal.pone.0017438
  26. 26. Møller HG, Rasmussen AP, Andersen HH, Johnsen KB, Henriksen M, Duroux M. A systematic review of microRNA in glioblastoma multiforme: Micro-modulators in the mesenchymal mode of migration and invasion. Molecular Neurobiology. 2013;47(1):131-144. DOI: 10.1007/s12035-012-8349-7
  27. 27. Aloizou AM, Pateraki G, Siokas V, Mentis AA, Liampas I, Lazopoulos G, et al. The role of MiRNA-21 in gliomas: Hope for a novel therapeutic intervention? Toxicology Reports. 2020;7:1514-1530. DOI: 10.1016/j.toxrep.2020.11.001
  28. 28. Yeh M, Wang YY, Yoo JY, Oh C, Otani Y, Kang JM, et al. MicroRNA-138 suppresses glioblastoma proliferation through downregulation of CD44. Scientific Reports. 2021;11(1):9219. DOI: 10.1038/s41598-021-88615-8
  29. 29. Figueroa J, Phillips LM, Shahar T, Hossain A, Gumin J, Kim H, et al. Exosomes from glioma-associated mesenchymal stem cells increase the Tumorigenicity of glioma stem-like cells via transfer of miR-1587. Cancer Research. 2017;77(21):5808-5819. DOI: 10.1158/0008-5472.can-16-2524
  30. 30. Buruiană A, Florian IȘ, Florian AI, Timiș TL, Mihu CM, Miclăuș M, et al. The roles of miRNA in glioblastoma tumor cell communication: Diplomatic and aggressive negotiations. International Journal of Molecular Sciences. 2020;21(6):1950. DOI: 10.3390/ijms21061950
  31. 31. Huse JT, Brennan C, Hambardzumyan D, Wee B, Pena J, Rouhanifard SH, et al. The PTEN-regulating microRNA miR-26a is amplified in high-grade glioma and facilitates gliomagenesis in vivo. Genes & Development. 2009;23(11):1327-1337. DOI: 10.1101/gad.1777409
  32. 32. Lu F, Li C, Sun Y, Jia T, Li N, Li H. Upregulation of miR-1825 inhibits the progression of glioblastoma by suppressing CDK14 though Wnt/β-catenin signaling pathway. World Journal of Surgical Oncology. 2020;18(1):147. DOI: 10.1186/s12957-020-01927-3
  33. 33. Yan Y, Yan H, Wang Q , Zhang L, Liu Y, Yu H. MicroRNA 10a induces glioma tumorigenesis by targeting myotubularin-related protein 3 and regulating the Wnt/β-catenin signaling pathway. The FEBS Journal. 2019;286(13):2577-2592. DOI: 10.1111/febs.14824
  34. 34. Wang J, Chen C, Yan X, Wang P. The role of miR-382-5p in glioma cell proliferation, migration and invasion. Oncotargets and Therapy. 2019;12:4993-5002. DOI: 10.2147/ott.s196322
  35. 35. Wang Q , Li X, Zhu Y, Yang P. MicroRNA-16 suppresses epithelial-mesenchymal transition-related gene expression in human glioma. Molecular Medicine Reports. 2014;10(6):3310-3314. DOI: 10.3892/mmr.2014.2583
  36. 36. Hagemann C, Anacker J, Ernestus RI, Vince GH. A complete compilation of matrix metalloproteinase expression in human malignant gliomas. World Journal of Clinical Oncology. 2012;3(5):67-79. DOI: 10.5306/wjco.v3.i5.67
  37. 37. Wang X, Chen X, Sun L, Bi X, He H, Chen L, et al. MicroRNA-34a inhibits cell growth and migration in human glioma cells via MMP-9. Molecular Medicine Reports. 2019;20(1):57-64. DOI: 10.3892/mmr.2019.10233
  38. 38. Peng G, Liu Y, Yang C, Shen C. MicroRNA-25 promotes cell proliferation, migration and invasion in glioma by directly targeting cell adhesion molecule 2. Experimental and Therapeutic Medicine. 2022;23(1):16. DOI: 10.3892/etm.2021.10938
  39. 39. Li Y, Wang Y, Yu L, Sun C, Cheng D, Yu S, et al. miR-146b-5p inhibits glioma migration and invasion by targeting MMP16. Cancer Letters. 2013;339(2):260-269. DOI: 10.1016/j.canlet.2013.06.018
  40. 40. Han J, Alvarez-Breckenridge CA, Wang QE, Yu J. TGF-β signaling and its targeting for glioma treatment. American Journal of Cancer Research. 2015;5(3):945-955
  41. 41. Wang ZH, Zhang QS, Duan YL, Zhang JL, Li GF, Zheng DL. TGF-β induced miR-132 enhances the activation of TGF-β signaling through inhibiting SMAD7 expression in glioma cells. Biochemical and Biophysical Research Communications. 2015;463(3):187-192. DOI: 10.1016/j.bbrc.2015.05.001
  42. 42. Li H, Li J, Chen L, Qi S, Yu S, Weng Z, et al. HERC3-mediated SMAD7 ubiquitination degradation promotes autophagy-induced EMT and Chemoresistance in glioblastoma. Clinical Cancer Research. 2019;25(12):3602-3616. DOI: 10.1158/1078-0432.ccr-18-3791
  43. 43. Rosińska S, Gavard J. Tumor vessels fuel the fire in glioblastoma. International Journal of Molecular Sciences. 2021;22(12):6514. DOI: 10.3390/ijms22126514
  44. 44. Ahir BK, Engelhard HH, Lakka SS. Tumor development and angiogenesis in adult brain tumor: Glioblastoma. Molecular Neurobiology. 2020;57(5):2461-2478. DOI: 10.1007/s12035-020-01892-8
  45. 45. Würdinger T, Tannous BA, Saydam O, Skog J, Grau S, Soutschek J, et al. miR-296 regulates growth factor receptor overexpression in angiogenic endothelial cells. Cancer Cell. 2008;14(5):382-393. DOI: 10.1016/j.ccr.2008.10.005
  46. 46. Dai D, Huang W, Lu Q , Chen H, Liu J, Hong B. miR-24 regulates angiogenesis in gliomas. Molecular Medicine Reports. 2018;18(1):358-368. DOI: 10.3892/mmr.2018.8978
  47. 47. Smits M, Nilsson J, Mir SE, van der Stoop PM, Hulleman E, Niers JM, et al. Würdinger: miR-101 is down-regulated in glioblastoma resulting in EZH2-induced proliferation, migration, and angiogenesis. Oncotarget. 2010;1(8):710-720. DOI: 10.18632/oncotarget.205
  48. 48. Chen X, Yang F, Zhang T, Wang W, Xi W, Li Y, et al. MiR-9 promotes tumorigenesis and angiogenesis and is activated by MYC and OCT4 in human glioma. Journal of Experimental & Clinical Cancer Research. 2019;38(1):99. DOI: 10.1186/s13046-019-1078-2
  49. 49. Lucero R, Zappulli V, Sammarco A, Murillo OD, Cheah PS, Srinivasan S, et al. Glioma-derived miRNA-containing extracellular vesicles induce angiogenesis by reprogramming brain endothelial cells. Cell Reports. 2020;30(7):2065-2074.e4. DOI: 10.1016/j.celrep.2020.01.073
  50. 50. Svensson A, Özen I, Genové G, Paul G, Bengzon J. Endogenous brain pericytes are widely activated and contribute to mouse glioma microvasculature. PLoS One. 2015;10(4):e0123553. DOI: 10.1371/journal.pone.0123553
  51. 51. Kim K, Kim IK, Yang JM, Lee E, Koh BI, Song S, et al. SoxF transcription factors are positive feedback regulators of VEGF signaling. Circulation Research. 2016;119(7):839-852. DOI: 10.1161/circresaha.116.308483
  52. 52. Wong HA, Fatimy RE, Onodera C, Wei Z, Yi M, Mohan A, et al. The cancer genome atlas analysis predicts MicroRNA for targeting cancer growth and vascularization in glioblastoma. Molecular Therapy. 2015;23(7):1234-1247. DOI: 10.1038/mt.2015.72
  53. 53. Wang ZF, Liao F, Wu H, Dai J. Glioma stem cells-derived exosomal miR-26a promotes angiogenesis of microvessel endothelial cells in glioma. Journal of Experimental & Clinical Cancer Research. 2019;38(1):201. DOI: 10.1186/s13046-019-1181-4
  54. 54. Yuan M, Da Silva A, Arnold A, Okeke L, Ames H, Correa-Cerro LS, et al. MicroRNA (miR) 125b regulates cell growth and invasion in pediatric low grade glioma. Scientific Reports. 2018;8(1):12506. DOI: 10.1038/s41598-018-30942-4
  55. 55. Smits M, Wurdinger T, van het Hof B, Drexhage JA, Geerts D, Wesseling P, et al. Myc-associated zinc finger protein (MAZ) is regulated by miR-125b and mediates VEGF-induced angiogenesis in glioblastoma. The FASEB Journal. 2012;26(6):2639-2647. DOI: 10.1096/fj.11-202820
  56. 56. Zhang G, Chen L, Khan AA, Li B, Gu B, Lin F, et al. miRNA-124-3p/neuropilin-1(NRP-1) axis plays an important role in mediating glioblastoma growth and angiogenesis. International Journal of Cancer. 2018;143(3):635-644. DOI: 10.1002/ijc.31329
  57. 57. Zheng X, Chopp M, Lu Y, Buller B, Jiang F. MiR-15b and miR-152 reduce glioma cell invasion and angiogenesis via NRP-2 and MMP-3. Cancer Letters. 2013;329(2):146-154. DOI: 10.1016/j.canlet.2012.10.026
  58. 58. Wang L, Lang B, Zhou Y, Ma J, Hu K. Up-regulation of miR-663a inhibits the cancer stem cell-like properties of glioma via repressing the KDM2A-mediated TGF-β/SMAD signaling pathway. Cell Cycle. 2021;20(19):1935-1952. DOI: 10.1080/15384101.2021.1966962
  59. 59. Korać P, Antica M, Matulić M. MiR-7 in cancer development. Biomedicine. 2021;9(3):325. DOI: 10.3390/biomedicines9030325
  60. 60. Geng L, Xu J, Zhu Y, Hu X, Liu Y, Yang K, et al. Targeting miR-9 in glioma stem cell-derived extracellular vesicles: A novel diagnostic and therapeutic biomarker. Translational Oncology. 2022;22:101451. DOI: 10.1016/j.tranon.2022.101451
  61. 61. Liu A, Zhao H, Sun B, Han X, Zhou D, Cui Z, et al. A predictive analysis approach for paediatric and adult high-grade glioma: miRNAs and network insight. The Annals of Translational Medicine. 2020;8(5):242. DOI: 10.21037/atm.2020.01.12
  62. 62. Karsy M, Arslan E, Moy F. Current Progress on understanding MicroRNAs in glioblastoma Multiforme. Genes & Cancer. 2012;3(1):3-15. DOI: 10.1177/1947601912448068
  63. 63. Cortez MA, Nicoloso MS, Shimizu M, Rossi S, Gopisetty G, Molina JR, et al. miR-29b and miR-125a regulate podoplanin and suppress invasion in glioblastoma. Genes, Chromosomes & Cancer. 2010;49(11):981-990. DOI: 10.1002/gcc.20808
  64. 64. Xia HF, He TZ, Liu CM, Cui Y, Song PP, Jin XH, et al. MiR-125b expression affects the proliferation and apoptosis of human glioma cells by targeting Bmf. Cellular Physiology and Biochemistry. 2009;23(4-6):347-358. DOI: 10.1159/000218181
  65. 65. Macharia LW, Muriithi W, Nyaga DK, de Mattos Coelho-Aguiar J. De Sampaio TCL. e Spohr and V Moura Neto: Evaluation of MiRNA expression in glioblastoma stem-like cells: A comparison between Normoxia and hypoxia microenvironment. Onco. 2022;2(2):113-128
  66. 66. Qian C, Wang B, Zou Y, Zhang Y, Hu X, Sun W, et al. MicroRNA 145 enhances chemosensitivity of glioblastoma stem cells to demethoxycurcumin. Cancer Management and Research. 2019;11:6829-6840. DOI: 10.2147/cmar.s210076
  67. 67. Zhang Z, Xu J, Chen Z, Wang H, Xue H, Yang C, et al. Transfer of MicroRNA via macrophage-derived extracellular vesicles promotes proneural-to-mesenchymal transition in glioma stem cells. Cancer Immunology Research. 2020;8(7):966-981. DOI: 10.1158/2326-6066.cir-19-0759
  68. 68. Schraivogel D, Weinmann L, Beier D, Tabatabai G, Eichner A, Zhu JY, et al. CAMTA1 is a novel tumour suppressor regulated by miR-9/9* in glioblastoma stem cells. The EMBO Journal. 2011;30(20):4309-4322. DOI: 10.1038/emboj.2011.301
  69. 69. Mahinfar P, Mansoori B, Rostamzadeh D, Baradaran B, Cho WC, Mansoori B. The role of microRNAs in multidrug resistance of glioblastoma. Cancers (Basel). 2022;14(13):3217. DOI: 10.3390/cancers14133217
  70. 70. Müller P, Abdel Gaber SA, Zimmermann W, Wittig R, Stepp H. ABCG2 influence on the efficiency of photodynamic therapy in glioblastoma cells. Journal of Photochemistry and Photobiology. B. 2020;210:111963. DOI: 10.1016/j.jphotobiol.2020.111963
  71. 71. Li WQ , Li YM, Tao BB, Lu YC, Hu GH, Liu HM, et al. Yu: Downregulation of ABCG2 expression in glioblastoma cancer stem cells with miRNA-328 may decrease their chemoresistance. Medical Science Monitor. 2010;16(10):Hy27-30
  72. 72. Wu Z, Sun L, Wang H, Yao J, Jiang C, Xu W, et al. MiR-328 expression is decreased in high-grade gliomas and is associated with worse survival in primary glioblastoma. PLoS One. 2012;7(10):e47270. DOI: 10.1371/journal.pone.0047270
  73. 73. Li Y, Liu Y, Ren J, Deng S, Yi G, Guo M, et al. miR-1268a regulates ABCC1 expression to mediate temozolomide resistance in glioblastoma. Journal of Neuro-Oncology. 2018;138(3):499-508. DOI: 10.1007/s11060-018-2835-3
  74. 74. Xu J, Song J, Xiao M, Wang C, Zhang Q , Yuan X, et al. RUNX1 (RUNX family transcription factor 1), a target of microRNA miR-128-3p, promotes temozolomide resistance in glioblastoma multiform by upregulating multidrug resistance-associated protein 1 (MRP1). Bioengineered. 2021;12(2):11768-11781. DOI: 10.1080/21655979.2021.2009976
  75. 75. Jeon HM, Sohn YW, Oh SY, Kim SH, Beck S, Kim S, et al. ID4 imparts chemoresistance and cancer stemness to glioma cells by derepressing miR-9*-mediated suppression of SOX2. Cancer Research. 2011;71(9):3410-3421. DOI: 10.1158/0008-5472.can-10-3340
  76. 76. Wang Z, Yang J, Xu G, Wang W, Liu C, Yang H, et al. Targeting miR-381-NEFL axis sensitizes glioblastoma cells to temozolomide by regulating stemness factors and multidrug resistance factors. Oncotarget. 2015;6(5):3147-3164. DOI: 10.18632/oncotarget.3061
  77. 77. Medarova Z, Pantazopoulos P, Yoo B. Screening of potential miRNA therapeutics for the prevention of multi-drug resistance in cancer cells. Scientific Reports. 2020;10(1):1970. DOI: 10.1038/s41598-020-58919-2
  78. 78. Strobel H, Baisch T, Fitzel R, Schilberg K, Siegelin MD, Karpel-Massler G, et al. Temozolomide and other alkylating agents in glioblastoma therapy. Biomedicine. 2019;7(3):69. DOI: 10.3390/biomedicines7030069
  79. 79. Hu Z, Mi Y, Qian H, Guo N, Yan A, Zhang Y, et al. A potential mechanism of Temozolomide resistance in glioma-Ferroptosis. Frontiers in Oncology. 2020;10:897. DOI: 10.3389/fonc.2020.00897
  80. 80. Ortiz R, Perazzoli G, Cabeza L, Jiménez-Luna C, Luque R, Prados J, et al. Temozolomide: An updated overview of resistance mechanisms, nanotechnology advances and clinical applications. Current Neuropharmacology. 2021;19(4):513-537. DOI: 10.2174/1570159x18666200626204005
  81. 81. Caccese M, Ius T, Simonelli M, Fassan M, Cesselli D, Dipasquale A, et al. Mismatch-repair protein expression in high-grade gliomas: A large retrospective multicenter study. International Journal of Molecular Sciences. 2020;21(18):6716. DOI: 10.3390/ijms21186716
  82. 82. Cheng YW, Lin CJ, Kuo SH, Lieu AS, Chai CY, Tsai HP, et al. miR-3059-3p regulates glioblastoma Multiforme Radiosensitivity enhancement through the homologous recombination pathway of DNA repair. Journal of Oncology. 2022;2022:7250278. DOI: 10.1155/2022/7250278
  83. 83. Li Q , Wang C, Cai L, Lu J, Zhu Z, Wang C, et al. miR-34a derived from mesenchymal stem cells stimulates senescence in glioma cells by inducing DNA damage. Molecular Medicine Reports. 2019;19(3):1849-1857. DOI: 10.3892/mmr.2018.9800
  84. 84. Yang L, Ma Y, Xin Y, Han R, Li R, Hao X. Role of the microRNA 181 family in glioma development. Molecular Medicine Reports. 2018;17(1):322-329. DOI: 10.3892/mmr.2017.7895
  85. 85. Nie E, Jin X, Wu W, Yu T, Zhou X, Shi Z, et al. MiR-198 enhances temozolomide sensitivity in glioblastoma by targeting MGMT. Journal of Neuro-Oncology. 2017;133(1):59-68. DOI: 10.1007/s11060-017-2425-9
  86. 86. Gao YT, Chen XB, Liu HL. Up-regulation of miR-370-3p restores glioblastoma multiforme sensitivity to temozolomide by influencing MGMT expression. Scientific Reports. 2016;6:32972. DOI: 10.1038/srep32972
  87. 87. Bandey I, Chiou SH, Huang AP, Tsai JC, Tu PH. Progranulin promotes Temozolomide resistance of glioblastoma by orchestrating DNA repair and tumor stemness. Oncogene. 2015;34(14):1853-1864. DOI: 10.1038/onc.2014.92
  88. 88. Zeng A, Wei Z, Yan W, Yin J, Huang X, Zhou X, et al. Exosomal transfer of miR-151a enhances chemosensitivity to temozolomide in drug-resistant glioblastoma. Cancer Letters. 2018;436:10-21. DOI: 10.1016/j.canlet.2018.08.004
  89. 89. Filipów S, Łaczmański Ł. Blood circulating miRNAs as cancer biomarkers for diagnosis and surgical treatment response. Frontiers in Genetics. 2019;10:169. DOI: 10.3389/fgene.2019.00169
  90. 90. Conti A, Aguennouz M, La Torre D, Tomasello C, Cardali S, Angileri FF, et al. miR-21 and 221 upregulation and miR-181b downregulation in human grade II-IV astrocytic tumors. Journal of Neuro-Oncology. 2009;93(3):325-332. DOI: 10.1007/s11060-009-9797-4
  91. 91. Akers JC, Ramakrishnan V, Kim R, Phillips S, Kaimal V, Mao Y, et al. miRNA contents of cerebrospinal fluid extracellular vesicles in glioblastoma patients. Journal of Neuro-Oncology. 2015;123(2):205-216. DOI: 10.1007/s11060-015-1784-3
  92. 92. Teplyuk NM, Mollenhauer B, Gabriely G, Giese A, Kim E, Smolsky M, et al. MicroRNAs in cerebrospinal fluid identify glioblastoma and metastatic brain cancers and reflect disease activity. Neuro-Oncology. 2012;14(6):689-700. DOI: 10.1093/neuonc/nos074
  93. 93. Wang Q , Li P, Li A, Jiang W, Wang H, Wang J, et al. Plasma specific miRNAs as predictive biomarkers for diagnosis and prognosis of glioma. Journal of Experimental & Clinical Cancer Research. 2012;31(1):97. DOI: 10.1186/1756-9966-31-97
  94. 94. Godlewski J, Krichevsky AM, Johnson MD, Chiocca EA, Bronisz A. Belonging to a network--microRNAs, extracellular vesicles, and the glioblastoma microenvironment. Neuro-Oncology. 2015;17(5):652-662. DOI: 10.1093/neuonc/nou292
  95. 95. Dong L, Li Y, Han C, Wang X, She L, Zhang H. miRNA microarray reveals specific expression in the peripheral blood of glioblastoma patients. International Journal of Oncology. 2014;45(2):746-756. DOI: 10.3892/ijo.2014.2459
  96. 96. van der Vos KE, Abels ER, Zhang X, Lai C, Carrizosa E, Oakley D, et al. Directly visualized glioblastoma-derived extracellular vesicles transfer RNA to microglia/macrophages in the brain. Neuro-Oncology. 2016;18(1):58-69. DOI: 10.1093/neuonc/nov244
  97. 97. Li E, Zhang J, Yuan T, Ma B. MiR-143 suppresses osteogenic differentiation by targeting Osterix. Molecular and Cellular Biochemistry. 2014;390(1-2):69-74. DOI: 10.1007/s11010-013-1957-3
  98. 98. Quirico L, Orso F. The power of microRNAs as diagnostic and prognostic biomarkers in liquid biopsies. Cancer Drug Resistance. 2020;3(2):117-139. DOI: 10.20517/cdr.2019.103
  99. 99. Rivera-Díaz M, Miranda-Román MA, Soto D, Quintero-Aguilo M, Ortiz-Zuazaga H, Marcos-Martinez MJ, et al. MicroRNA-27a distinguishes glioblastoma multiforme from diffuse and anaplastic astrocytomas and has prognostic value. American Journal of Cancer Research. 2015;5(1):201-218
  100. 100. Huang SW, Ali ND, Zhong L, Shi J. MicroRNAs as biomarkers for human glioblastoma: Progress and potential. Acta Pharmacologica Sinica. 2018;39(9):1405-1413. DOI: 10.1038/aps.2017.173
  101. 101. Akers JC, Hua W, Li H, Ramakrishnan V, Yang Z, Quan K, et al. A cerebrospinal fluid microRNA signature as biomarker for glioblastoma. Oncotarget. 2017;8(40):68769-68779. DOI: 10.18632/oncotarget.18332
  102. 102. Wang J, Che F, Zhang J. Cell-free microRNAs as non-invasive biomarkers in glioma: A diagnostic meta-analysis. The International Journal of Biological Markers. 2019;34(3):232-242. DOI: 10.1177/1724600819840033
  103. 103. Kim TM, Huang W, Park R, Park PJ, Johnson MD. A developmental taxonomy of glioblastoma defined and maintained by MicroRNAs. Cancer Research. 2011;71(9):3387-3399. DOI: 10.1158/0008-5472.can-10-4117
  104. 104. Ye Z, Zhang Z, Wu L, Liu C, Chen Q , Liu J, et al. Upregulation of miR-183 expression and its clinical significance in human brain glioma. Neurological Sciences. 2016;37(8):1341-1347. DOI: 10.1007/s10072-016-2599-5
  105. 105. Arghiani N, Shah K. Modulating microRNAs in cancer: Next-generation therapies. Cancer Biology & Medicine. 2021;19(3):289-304. DOI: 10.20892/j.issn.2095-3941.2021.0294
  106. 106. Cho WC. OncomiRs: The discovery and progress of microRNAs in cancers. Molecular Cancer. 2007;6:60. DOI: 10.1186/1476-4598-6-60
  107. 107. Bader AG, Brown D, Winkler M. The promise of microRNA replacement therapy. Cancer Research. 2010;70(18):7027-7030. DOI: 10.1158/0008-5472.can-10-2010
  108. 108. Bhat SA, Majid S, Rehman MU. Scenario and future prospects of microRNAs in gastric cancer: A review. Iranian Journal of Basic Medical Sciences. 2019;22(4):345-352. DOI: 10.22038/ijbms.2019.32399.7765
  109. 109. Ananta JS, Paulmurugan R, Massoud TF. Nanoparticle-delivered antisense MicroRNA-21 enhances the effects of Temozolomide on glioblastoma cells. Molecular Pharmaceutics. 2015;12(12):4509-4517. DOI: 10.1021/acs.molpharmaceut.5b00694
  110. 110. Ananta JS, Paulmurugan R, Massoud TF. Tailored nanoparticle Codelivery of antimiR-21 and antimiR-10b augments glioblastoma cell kill by Temozolomide: Toward a "personalized" anti-microRNA therapy. Molecular Pharmaceutics. 2016;13(9):3164-3175. DOI: 10.1021/acs.molpharmaceut.6b00388
  111. 111. Malhotra M, Sekar TV, Ananta JS, Devulapally R, Afjei R, Babikir HA, et al. Targeted nanoparticle delivery of therapeutic antisense microRNAs presensitizes glioblastoma cells to lower effective doses of temozolomide in vitro and in a mouse model. Oncotarget. 2018;9(30):21478-21494. DOI: 10.18632/oncotarget.25135
  112. 112. Chen Y, Gao DY, Huang L. In vivo delivery of miRNAs for cancer therapy: Challenges and strategies. Advanced Drug Delivery Reviews. 2015;81:128-141. DOI: 10.1016/j.addr.2014.05.009

Written By

Manendra Singh Tomar and Ashutosh Shrivastava

Submitted: 06 June 2023 Reviewed: 06 June 2023 Published: 29 June 2023