Skip to main content

Exploring the clinical implications and applications of exosomal miRNAs in gliomas: a comprehensive study

Abstract

Gliomas are aggressive brain tumors associated with poor prognosis and limited treatment options due to their invasive nature and resistance to current therapeutic modalities. Research suggests that exosomal microRNAs have emerged as key players in intercellular communication within the tumor microenvironment, influencing tumor progression and therapeutic responses. Exosomal microRNAs (miRNAs), small non-coding RNAs, are crucial in glioma development, invasion, metastasis, angiogenesis, and immune evasion by binding to target genes. This comprehensive review examines the clinical relevance and implications of exosomal miRNAs in gliomas, highlighting their potential as diagnostic biomarkers, therapeutic targets and prognosis biomarker. Additionally, we also discuss the limitations of current exsomal miRNA treatments and address challenges and propose future directions for leveraging exosomal miRNAs in precision oncology for glioma management.

Introduction

The World Health Organization (WHO) CNS5 classification of central nervous system tumors categorizes gliomas into grades I to IV, with higher grades indicating higher malignancy [1,2,3]. Grades I and II gliomas are classified as low-grade gliomas, while grades III and IV are classified as high-grade gliomas. Low-grade brain gliomas primarily refer to diffuse astrocytomas, oligodendrogliomas, and oligoastrocytomas. High-grade brain gliomas primarily refer to anaplastic astrocytomas, anaplastic oligodendrogliomas, glioblastomas, and diffuse midline gliomas. Gliomas, the most common and malignant primary brain neoplasms, originate from glial or precursor cells in the central nervous system (CNS) neuroectoderm [4, 5]. They are characterized by their undesirable prognosis and poor survival rate, encompassing astrocytomas, oligodendrogliomas, and ependymomas [6, 7], posing a serious threat to human health and lives. While computed tomography (CT) and magnetic resonance imaging (MRI) are routine techniques for diagnosing disease stages and monitoring tumor growth and therapeutic response, they are often cumbersome and expensive. Conventional surgical, radiation, and chemotherapy treatments can delay tumor progression but are often ineffective and do not provide significant improvement [8]. Therefore, there is an urgent need for a new method that can both diagnose and treat gliomas, as well as detect the prognosis of gliomas. Recently, increasing evidence suggests that diagnostic biomarkers, such as exosomal miRNAs, would be clinically meaningful for the early detection of the tumor and for cases in which surgery is contraindicated or biopsy results are inconclusive [9, 10].

Exosomes, small vesicles with round or cup shapes, were first isolated from sheep erythrocyte supernatant in 1983 by JohnStone et al. during a study on the transformation of immature erythrocytes to mature erythrocytes [11]. These vesicles, which have a diameter of 40–160 nm, originate from endosomes and consist of lipid bilayer membranes [12, 13]. They are actively released by most cells, circulating stably in body fluids [14, 15]. Besides their roles in cell-to-cell communication and tumorigenesis, exosomes safeguard the substances they carry, including DNA, RNA, lipids, and proteins, from degradation [16,17,18,19,20], while also regulating the activity of recipient cells and influencing the tumor microenvironment by transporting nucleic acids [21].

Exosomal miRNAs, a subset of circulating miRNAs, have gained attention for their characteristics as endogenous, double-stranded, non-coding small molecule RNAs, typically 18–25 nucleotides in length, with over 1000 different exosomal miRNAs identified through RNA sequencing [22, 23]. The discovery of the first miRNA dates back to 1993 when Lee and Ambros identified one in Caenorhabditis elegans, and their presence in exosomes was first demonstrated by Valadi et al. in 2007 [24,25,26,27]. Originating from single-stranded miRNA gene transcripts, they are processed by the ribonuclease III enzyme, Dicer, integrated into the RNA-induced silencing complex, enabling them to repress the translation of target RNA by binding to partially complementary sequences in the 3ʹ untranslated region (UTR) of messenger RNA (mRNA) [28,29,30] (Fig. 1). Exosomal miRNAs may play roles in gene activation in certain contexts, affecting biological processes, exerting significant influence on cell differentiation, proliferation, and survival, exhibiting both tumor-suppressive and oncogenic effects [31,32,33,34]. Notably, Calin et al. observed a loss or down-regulation of miR-15/16 cluster expression in chronic lymphocytic leukemia in 2002, highlighting the potential role of miRNAs in tumorigenesis [35]. Studies have also demonstrated the effectiveness of exosomal miRNAs as potential biomarkers in liquid biopsy for cancer diagnosis, treatment monitoring, and prognosis prediction, emerging as valuable disease markers [36,37,38,39,40,41,42,43]. Notably, the effects of exosomal miRNAs on gliomas primarily involve the inhibition or promotion of their downstream target genes, thereby exerting their respective biological functions (Tables 1, 2).

Fig. 1
figure 1

The process of production and functions of mature exosomal miRNAs in animals. Mature miRNA is secreted out of the cells after entering endosomes formed by the cell membrane, thus resulting in the formation of exosomal miRNA. Mature miRNAs are produced in two stages: in the nucleus, genes encoding miRNAs are transcribed into pri-miRNAs by the action of RNA polymerase RNA pol II, which are then processed by Drosha RNase III nucleic acid endonuclease into 60-70 nt stem-loop intermediates, i.e. pre-miRNAs (miRNA precursors). Subsequently, pre-miRNA is transported into the cytoplasm by Exportin-5 bound to Ran-GTP, and the stem-loop structure is cleaved away by Dicer RNase III nuclease, leaving two incompletely paired strands called miRNA:miRNA* complexes, where miRNA is the mature miRNA and miRNA* is the relative arm with a short lifetime. Then, the double strand is loaded onto the argonaute protein, and nucleotide base pairing between the miRNA in the double strand and the complementary sequence in the 3′ untranslated region (3′ UTR) of the target mRNA forms the miRNA effector miRNA-induced silencing complex (miRISC) to inhibit translation and/or promote mRNA degradation, while miRNAs are released and degraded

Table 1 Exosomal miRNAs that are down-regulated, their target genes and biological functions, and their roles in glioma diagnosis, treatment and prognosis
Table 2 Exosomal miRNAs that are upregulated, their target genes and biological functions, and their roles in glioma diagnosis, treatment and prognosis

In this review, we summarize the latest findings on the fundamental roles of exosomal miRNAs implicated in the diagnosis, treatment, and prognosis of gliomas, considering the complex molecular mechanisms and the expanding research field. We also discuss the research limitations and provide future perspectives.

Exosomal miRNAs and the occurrence and development of gliomas

Exosomal miRNA biogenesis is tightly regulated temporally and spatially, with dysregulation implicated in glioma development [44,45,46,47,48,49]. Current research suggests that dysregulation of exosomal miRNAs could be pivotal in glioma growth, angiogenesis, metastasis, and cell migration. For example, several studies have reported that miR-375 inhibits proliferation and invasion in glioblastoma, while miR-16-1 inhibits migration and proliferation in glioma cells [50, 51]. Additionally, Li et al. demonstrated that microglial exosome miR-7239-3p promotes glioma progression by regulating Circadian genes [52]. These research findings indicate a close association between exosomal miRNAs and the occurrence and development of gliomas, suggesting their potential application in clinical diagnosis, treatment, and prognosis.

Application of exosomal miRNAs in the diagnosis of gliomas

A variety of circulating biomarkers are present in the blood of glioma patients, including circulating tumor cells, nucleic acids, and proteins [53]. Among these, exosomal miRNAs have garnered considerable attention due to their significant distinctions between glioma patients and healthy counterparts. Lu et al. conducted a systematic analysis of 217 mammalian miRNAs across 334 samples, revealing a prevailing downregulation of miRNAs in tumors compared to normal tissues [54]. Their findings enabled the successful classification of poorly differentiated tumors using miRNA expression profiling, highlighting the diagnostic potential of miRNA analysis in cancer. Furthermore, Wang et al. discovered significantly higher levels of miR-766-5p and miR-376b-5p in the serum of patients with high-grade glioma compared to healthy controls, suggesting their potential as auxiliary diagnostic biomarkers [55].

Glioblastoma (GBM), or glioblastoma multiforme, is the most malignant and common type of glioma. It is classified as a Grade IV glioma, characterized by its rapid growth, high invasiveness, and poor prognosis. Dong et al. identified elevated expressions of miR-576-5p, miR-340, and miR-626, alongside decreased expressions of miR-320, miR-7-5p, and let-7g-5p in the peripheral blood of GBM patients compared to normal subjects using gene microarray analysis [56]. In another study by Manterola, analysis of exosomal miRNAs from 161 GBM patients and 110 healthy subjects revealed that the combination of miR-320/miR-574-3p/RNU6-1 (RNA, U6 Small Nuclear 1) serves as promising biomarker candidates for distinguishing between GBM patients and healthy controls [57]. Similarly, elevated levels of miR-103 and miR-125 were detected in the serum of GBM patients [58].

Numerous studies have reported a significant upregulation of exosomal miR-21 expression in the serum of glioma patients, a dysregulation that may contribute to the initiation and progression of GBM by influencing various cellular and molecular targets [59, 60]. Santangelo et al. observed that post-surgery, levels of exosomal miR-21 isolated from patient serum were notably higher in individuals with high-grade glioma compared to those with low-grade glioma [61]. They further identified three serum exosome-associated miRNAs (miR-21, miR-222, miR-124-3) that could aid in glioma detection and grading through the analysis of 141 serum samples and accompanying clinical data [40]. The utilization of these three miRNA-based diagnostics was demonstrated to enhance diagnostic efficiency for high-grade glioma patients. Additionally, Akers et al. noted significantly elevated levels of miR-21 in the cerebrospinal fluid of GBM patients during their examination of cerebrospinal fluid from both healthy subjects and GBM patients for miRNA analysis [62]. Consequently, exosomal miR-21 emerges as a reliable biomarker for the diagnosis of glioma patients.

In addition to exosomal miR-21, several exosomal miRNAs have shown potential as biomarkers for glioma diagnosis. MiR-10, which is absent in normal brain tissue but elevated in gliomas, has been linked to tumor cell stasis, apoptosis, and autophagy [63]. MiR-449 and miR-5194 are promising for GBM diagnosis, while miR-210 is already confirmed as a biomarker for GBM patients [64]. Additionally, miR-221 was found to be significantly upregulated in GBM tissues, while miR-128 and miR-181 expressions were decreased, suggesting that elevated miR-221 expression could serve as a potential diagnostic biomarker for glioma [65, 66].

Tumor metastasis is a critical stage of tumor progression and a major therapeutic challenge. Exosomal miRNAs play an important role in the development of distant metastasis of primary tumor cells [67]. Studies have demonstrated a significant elevation in miR-148a levels in both the serum and tumor tissues of GBM patients. Furthermore, miR-148a has been implicated in promoting glioma metastasis, proliferation, and migration by targeting Cell Adhesion Molecule 1(CADM1), Mitogen-inducible gene 6(MIG6), and Epidermal growth factor receptor (EGFR) [68, 69]. Additionally, Teplyuk et al. also observed significant elevations in miR-10b and miR-21 levels in the cerebrospinal fluid of GBM patients, whereas miR-200 levels were notably increased in the cerebrospinal fluid of patients with glioma metastases [70]. Furthermore, Emilliya and colleagues discovered that miR-491 not only differentiates glioma brain metastasis but also exhibits lower expression in high-grade gliomas compared to low-grade gliomas [71]. Meanwhile, the findings of Bao et al. indicate that elevated expression of miR-155-5p in the plasma of glioma patients is positively correlated with glioma grading [72]. These studies suggest that miRNAs in exosomes have the potential to serve as diagnostic markers for glioma metastasis and to distinguish between different grades of gliomas, such as low-grade and high-grade gliomas.

Application of exosomal miRNAs for the treatment of gliomas

Traditional glioma treatments, like surgical resection and chemotherapy with radiotherapy, often lead to significant side effects and poor prognosis [73, 74]. For example, temozolomide (TMZ), a primary antitumor agent in clinical chemotherapy, induces DNA damage in glioma cells while also causing a multitude of chemotherapy side effects [75]. Furthermore, the limitations of traditional treatments underscore the urgent need for alternative therapeutic approaches. Nucleic acid therapy, currently promising in various human diseases, has attracted attention. Research accumulation indicates potential utilization of exosomal miRNAs in treating malignant tumors. [76, 77]. These exosomal miRNAs regulate gene expression post-transcriptionally and intricately connect with the glioma microenvironment through targeting multiple signaling pathways such as EGFR, Phosphatidylinositide 3-kinases/protein kinase B (PI3K/AKT), p53, Notch, and others [78,79,80,81].

Targeted therapy refers to a treatment strategy that specifically targets molecules or molecular pathways involved in the growth, progression, or spread of diseases such as cancer. Unlike traditional chemotherapy, which can affect both cancerous and healthy cells, targeted therapies are designed to interfere with specific molecules that play crucial roles in disease development. Meanwhile, exosomal miRNAs possess dual characteristics, with both tumor suppressive and oncogenic activities, endowing them with potential for tumor therapy [82,83,84]. Dysregulation of exosomal miRNAs significantly impacts glioma tumorigenesis, proliferation, migration, invasion, angiogenesis, immunosuppression, and drug resistance, suggesting a potentially innovative clinical treatment approach for glioma (Fig. 2).

Fig. 2
figure 2

The relationship between exosomal miRNAs, their target genes and biological functions. Exosomal miRNAs mediate their biological effects by targeting downstream genes, playing a pivotal role in shaping the glioma microenvironment. These effects include influencing glioma angiogenesis, progression, proliferation, invasion, migration, and the development of treatment resistance

Exosomal miRNAs for drug-resistant glioma treatment

Glioma resistance to clinical chemotherapy drugs often results in frequent disease recurrence and a poor prognosis [85]. Numerous studies have demonstrated the involvement of exosomal miRNAs in glioma chemotherapy drug resistance, including alkylating drug TMZ, suggesting a novel treatment approach to enhance drug sensitivity by regulating the expression of exosomal miRNAs [86].

Yin et al. identified high expression of miR-1238 in TMZ-resistant GBM cells and tissues, suggesting the miR-1238/CAV1 (Caveolin 1) axis as a potential target for future anti-tumor drugs [87]. Additionally, Wang et al. demonstrated that miR-25-3p promotes glioma development and resistance to TMZ by downregulating F-Box And WD Repeat Domain Containing 7(FBXW7) [88]. Furthermore, overexpression of miR-199a enhances chemosensitivity to TMZ and inhibits glioma progression by downregulating ArfGAP With GTPase Domain, Ankyrin Repeat And PH Domain 2(AGAP2) [89]. Moreover, Munoz et al. demonstrated that targeted delivery of exosome-derived functional anti-miR-9 from bone marrow stem cells enhances sensitivity of GBM to TMZ [90]. Meanwhile, Sharif et al. discovered that exogenous miR-124 delivered through Wharton’s Jelly-derived MSCs (WJ-MSCs) efficiently affects cell migration and proliferation in GBM cells, increasing sensitivity to TMZ, suggesting a potential combination therapy for GBM [91].

Although the above studies have shown that exosomal miRNAs can enhance the sensitivity of glioma to chemotherapy drugs, most experiments are currently limited to the cell level. Whether it has the same effect in living animals and clinical trials still requires further exploration and research.

Stem cell-derived exosomal miRNAs for the treatment of gliomas

Stem cell-derived exosomal miRNAs are pivotal in glioma, affecting tumor growth, drug resistance, and treatment outcome [92, 93].They regulate gene expression and cell behavior, influencing tumor progression and treatment outcomes. Ongoing research aims to pinpoint specific miRNAs, elucidate their mechanisms, and explore their potential as biomarkers and therapeutic targets for improved glioma management [94, 95].

Glioma stem cells (GSCs) play a pivotal role in GBM, with GSC-derived exosomes (GSC-Exs) shown to enhance endothelial cell angiogenic ability via the miR-21/VEGF/Vascular Endothelial Growth Factor(VEGFR2) signaling pathway [96]. Jiang et al. demonstrated that GSC-derived exosomal miR-944 suppresses Vascular Endothelial Growth Factor C(VEGFC) expression and the AKT/ERK signaling pathway, inhibiting glioma growth, progression, and angiogenesis, offering potential for GBM therapeutic targeting [97]. Additionally, targeting Roundabout Guidance Receptor 1(ROBO1), miR-29a-3p delivered via exosomes from engineered human mesenchymal stem cells suppresses tumor migration and vasculogenic mimicry in glioma, presenting potential for anti-VM (Vasculogenic mimicry) therapy and as supplements for anti-angiogenic therapy [98]. Kim et al. found that exosomal miRNA-584 inhibits glioma metastasis, suggesting MSC-derived exosomal miRNAs as an alternative strategy for malignant glioma treatment [99].

Although stem cell-derived exosomal miRNAs are closely related to the therapeutic application of glioma, further research is still needed to understand their precise mechanisms, ensure effective delivery, and verify safety. These include optimizing miRNA-based therapies, using advanced delivery systems, conducting rigorous preclinical and clinical evaluations, and combining these methods with existing treatment modalities to improve their efficacy and targeting accuracy.

Type II macrophage-derived exosomal miRNAs for the treatment of gliomas

It has been shown that M2-type macrophages promote glioma proliferation and migration [100, 101]. Yao et al. found that miR-15a and miR-92a, lowly expressed in M2 macrophage-derived exosomes, inhibit glioma metastasis and infiltration by binding to Cyclin D1(CCND1) and RAP1B (RAP1B, Member Of RAS Oncogene Family) to activate the PI3K/AKT/mTOR (mammalian target of rapamycin) signaling pathway [102]. This suggests that miR-15a and miR-92a might be a novel biomarker for GBM diagnosis in the glioma patients, and targeting miR-15a or miR-92a could contribute to anti-tumor immunotherapy.

At present, the specific mechanism of action of M2-type macrophage-derived exosomal miRNAs in glioma is not fully understood and has been less studied. At the same time, their detection methods need to improve sensitivity and specificity, and their biological functions need to be further verified. In addition, there are also verification and safety issues in the transformation process from the research stage to actual clinical application.

Cellular hypoxia-derived exosomal miRNAs for the treatment of gliomas

Myeloid-derived suppressor cells (MDSCs) constitute a heterogeneous population of bone marrow-derived cells crucial for tumor immune escape mechanisms due to their ability to significantly suppress immune cell responses [103]. Meanwhile, hypoxia has been reported to play a critical role in miRNA release from exosomes and the differentiation progression of MDSCs.

Gou et al. conducted miRNA sequencing, revealing upregulation of miR-10a and miR-21 expressions in Hypoxia-induced glioma exosomes (H-GDEs) compared to normoxia [104]. These miRNAs enhanced MDSC proliferation by targeting Rora and PTEN genes, respectively. Knockdown experiments of miR-10a and miR-21 in glioma cells further confirmed these findings. Guo et al. demonstrated that hypoxia-induced glioma cell exosomes stimulated the differentiation of functional MDSCs by transporting miR-29a and miR-92a, targeting Hbp1 and Prkar1a, respectively [105]. This promoted MDSC proliferation, thereby regulating the immunosuppressive tumor microenvironment.

Additionally, hypoxic glioma cells secrete exosomal miR-301a, which activates the Wnt/β-catenin signaling pathway and enhances radiation resistance by targeting Transcription Elongation Factor A Like 7(TCEAL7). The exo-miR-301a/TCEAL7-signaling axis presents a novel target for cellular resistance to cancer therapeutic radiation in GBM patients [106]. Furthermore, exosomal miR-1246 and miR-10b-5p from hypoxic glioma directly target Fyn Related Src Family Tyrosine Kinase(FRK) and Transcription Factor AP-2 Alpha(TFAP2A), respectively [107]. These miRNAs are delivered to normoxic glioma cells, promoting cell migration and invasion, thus offering a new avenue for antitumor therapy development.

Application of exosomal miRNAs in the prognosis of gliomas

Recent research highlights the crucial role of exosomal miRNAs in determining glioma prognosis. Their expression levels serve as potential biomarkers for predicting clinical outcomes and guiding therapeutic strategies.

For example, Guessous et al. analyzed TCGA data and found that miR-10b levels were negatively correlated with the prognosis of glioma patients. Shi et al. observed an association between elevated exosomal miR-21 levels in the cerebrospinal fluid of glioma patients and poor prognosis [108]. In addition, serum exosomal miR-301a levels were significantly elevated in glioma patients compared to healthy controls. After surgical resection of the primary tumor, serum exosomal miR-301a levels decreased, but increased again upon tumor recurrence [109]. Hence, serum adventitial miR-301a expression may serve as a novel potential biomarker for predicting prognosis in advanced glioma cases. Furthermore, Stakaitis et al. reported that high levels of miR-181b were linked to shorter postoperative survival, and that exosomal miR-181 levels decreased during glioma progression [110]. This suggests that exosomal miR-181 may serve as a potential biomarker for prognosis in glioma patients.

Studies have identified several exosomal miRNAs as potential prognostic markers for glioblastoma GBM. In GBM patients, an inverse relationship was observed between miR-125b/miR-182-5p and nestin expression, which correlated with overall survival and implied their utility as a potential biomarker for predicting GBM prognosis [111, 112]. High expression of miR-454-3p/miR-10b in exosomes or miR-628-3p downregulation in GBM patients' blood suggests its potential as a poor prognosis biomarker [113,114,115]. Zottel et al. found that GBM patients with high miR-5p and miR-138-5p expressions, particularly with Isocitrate dehydrogenase (IDH) mutations, had significantly shorter median survival and worse prognosis [116]. Additionally, Sun et al. observed that miR-2276-5p was significantly lower in GBM patients compared to non-glioma individuals, correlating with poorer survival, while its target gene RAB13 (RAB13, Member RAS Oncogene Family) was elevated and associated with worse outcomes [117]. Moreover, Qiu et al. conducted bioinformatics analysis on 480 GBM samples, revealing strong associations between high levels of miR-326/-130a and low levels of miR-323/329/155/210 with long overall survival and progression-free survival in GBM patients [118]. These findings suggest that these miRNAs could serve as valuable prognostic biomarkers for GBM.

Conclusion

Gliomas, a type of brain tumor, can be broadly classified into two categories: localized gliomas, which are typically benign and often amenable to complete removal through surgical resection, and diffuse gliomas, which are malignant and carry a generally poor prognosis, posing significant challenges in post-surgery treatment [119]. As medical science progresses, the pursuit of such innovative approaches becomes crucial in enhancing patient care and outcomes in glioma cases. Exosomal miRNAs, small non-coding RNAs pivotal in regulating gene expression, have emerged as key players in various biological processes, including the pathogenesis of cancers, notably gliomas [120, 121]. Exosomal miRNAs present promising prospects for glioma research and treatment, offering potential for early diagnosis and prognostic assessment through liquid biopsies [53]. However, challenges include potential immune responses, off-target effects, and cytotoxicity, which could impact normal cells. In addition, the financial costs are substantial, encompassing isolation, extraction, and detection expenses, with overall development and clinical trials potentially costing several million dollars. Despite these hurdles, their value in glioma diagnostics, treatment and prognosis underscores their significance in ongoing research and clinical applications.

As the same time, the integration of big data analysis in exosomal miRNAs detection offers a groundbreaking perspective on gliomas [122]. With rapid advancements in bioinformatics and computational biology, big data analytics enable a deeper understanding of the intricate roles of exosomal miRNAs in glioma, driving the progress of personalized medicine [123]. For example, distinct exosomal miRNA patterns could serve as diagnostic markers, allowing physicians to diagnose and subtype gliomas via blood tests without invasive surgeries [124, 125]. Furthermore, in-depth analysis of miRNA expression patterns can inform treatment decisions, offering patients more personalized therapy options.

Additionally, exosomal miRNAs are prepared using various techniques, including ultracentrifugation, density gradient centrifugation, and immunoaffinity capture. Extraction is typically performed with commercial RNA kits, and detection is conducted using methods such as qRT-PCR and next-generation sequencing [126, 127]. These miRNAs are promising as non-invasive biomarkers for early cancer detection and prognosis, including gliomas, due to their ability to reflect tumor biology and facilitate liquid biopsy [128, 129]. However, several challenges persist, including the lack of standardized methods, ensuring high specificity and sensitivity, validating biomarkers across diverse patient cohorts, and addressing issues related to delivery and safety [130]. Future research should aim to standardize protocols, improve detection technologies, validate biomarkers through extensive clinical trials, and advance exosome engineering to enhance therapeutic applications. Collaboration among researchers, clinicians, and industry stakeholders will be essential for overcoming these challenges and advancing personalized medicine.

Availability of data and materials

No datasets were generated or analysed during the current study.

References

  1. Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, Hawkins C, Ng HK, Pfister SM, Reifenberger G, Soffietti R, von Deimling A, Ellison DW. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol. 2021;23(2021):1231–51. https://doi.org/10.1093/neuonc/noab106.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P, Ellison DW. The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 2016;131(2016):803–20. https://doi.org/10.1007/s00401-016-1545-1.

    Article  PubMed  Google Scholar 

  3. Bettinger I, Thanos S, Paulus W. Microglia promote glioma migration. Acta Neuropathol. 2002;103:351–5. https://doi.org/10.1007/s00401-001-0472-x.

    Article  PubMed  Google Scholar 

  4. Sanai N, Berger MS. Surgical oncology for gliomas: the state of the art. Nat Rev Clin Oncol. 2018;15:112–25. https://doi.org/10.1038/nrclinonc.2017.171.

    Article  PubMed  Google Scholar 

  5. Yang K, Wu Z, Zhang H, Zhang N, Wu W, Wang Z, Dai Z, Zhang X, Zhang L, Peng Y, Ye W, Zeng W, Liu Z, Cheng Q. Glioma targeted therapy: insight into future of molecular approaches. Mol Cancer. 2022;21:39. https://doi.org/10.1186/s12943-022-01513-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Bi J, Chowdhry S, Wu S, Zhang W, Masui K, Mischel PS. Altered cellular metabolism in gliomas - an emerging landscape of actionable co-dependency targets. Nat Rev Cancer. 2020;20:57–70. https://doi.org/10.1038/s41568-019-0226-5.

    Article  CAS  PubMed  Google Scholar 

  7. Lapointe S, Perry A, Butowski NA. Primary brain tumours in adults. The Lancet. 2018;392:432–46. https://doi.org/10.1016/s0140-6736(18)30990-5.

    Article  Google Scholar 

  8. Tan AC, Ashley DM, Lopez GY, Malinzak M, Friedman HS, Khasraw M. Management of glioblastoma: state of the art and future directions. CA Cancer J Clin. 2020;70:299–312. https://doi.org/10.3322/caac.21613.

    Article  PubMed  Google Scholar 

  9. Westphal M, Lamszus K. Circulating biomarkers for gliomas. Nat Rev Neurol. 2015;11:556–66. https://doi.org/10.1038/nrneurol.2015.171.

    Article  CAS  PubMed  Google Scholar 

  10. Reifenberger G, Wirsching HG, Knobbe-Thomsen CB, Weller M. Advances in the molecular genetics of gliomas - implications for classification and therapy. Nat Rev Clin Oncol. 2017;14:434–52. https://doi.org/10.1038/nrclinonc.2016.204.

    Article  CAS  PubMed  Google Scholar 

  11. Johnstone RM, Bianchini A, Teng K. Reticulocyte maturation and exosome release: transferrin receptor containing exosomes shows multiple plasma membrane functions. Blood. 1989;74:1844–51. https://doi.org/10.1182/blood.V74.5.1844.1844.

    Article  CAS  PubMed  Google Scholar 

  12. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478):aau6977. https://doi.org/10.1126/science.aau6977.

    Article  CAS  Google Scholar 

  13. Isaac R, Reis FCG, Ying W, Olefsky JM. Exosomes as mediators of intercellular crosstalk in metabolism. Cell Metab. 2021;33:1744–62. https://doi.org/10.1016/j.cmet.2021.08.006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Caby MP, Lankar D, Vincendeau-Scherrer C, Raposo G, Bonnerot C. Exosomal-like vesicles are present in human blood plasma. Int Immunol. 2005;17:879–87. https://doi.org/10.1093/intimm/dxh267.

    Article  CAS  PubMed  Google Scholar 

  15. Bastos P, Ferreira R, Manadas B, Moreira PI, Vitorino R. Insights into the human brain proteome: disclosing the biological meaning of protein networks in cerebrospinal fluid. Crit Rev Clin Lab Sci. 2017;54:185–204. https://doi.org/10.1080/10408363.2017.1299682.

    Article  CAS  PubMed  Google Scholar 

  16. Matarredona ER, Pastor AM. Extracellular vesicle-mediated communication between the glioblastoma and its microenvironment. Cells. 2019;9(1):96. https://doi.org/10.3390/cells9010096.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Yu D, Li Y, Wang M, Gu J, Xu W, Cai H, Fang X, Zhang X. Exosomes as a new frontier of cancer liquid biopsy. Mol Cancer. 2022;21:56. https://doi.org/10.1186/s12943-022-01509-9.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Skotland T, Sandvig K, Llorente A. Lipids in exosomes: Current knowledge and the way forward. Prog Lipid Res. 2017;66:30–41. https://doi.org/10.1016/j.plipres.2017.03.001.

    Article  CAS  PubMed  Google Scholar 

  19. Kosaka N, Yoshioka Y, Fujita Y, Ochiya T. Versatile roles of extracellular vesicles in cancer. J Clin Invest. 2016;126:1163–72. https://doi.org/10.1172/JCI81130.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Kalluri R. The biology and function of exosomes in cancer. J Clin Invest. 2016;126:1208–15. https://doi.org/10.1172/JCI81135.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Bian EB, Chen EF, Xu YD, Yang ZH, Tang F, Ma CC, Wang HL, Zhao B. Exosomal lncRNA-ATB activates astrocytes that promote glioma cell invasion. Int J Oncol. 2019;54:713–21. https://doi.org/10.3892/ijo.2018.4644.

    Article  CAS  PubMed  Google Scholar 

  22. Kristensen LS, Andersen MS, Stagsted LVW, Ebbesen KK, Hansen TB, Kjems J. The biogenesis, biology and characterization of circular RNAs. Nat Rev Genet. 2019;20:675–91. https://doi.org/10.1038/s41576-019-0158-7.

    Article  CAS  PubMed  Google Scholar 

  23. Goodall GJ, Wickramasinghe VO. RNA in cancer. Nat Rev Cancer. 2021;21:22–36. https://doi.org/10.1038/s41568-020-00306-0.

    Article  CAS  PubMed  Google Scholar 

  24. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75:843–54.

    Article  CAS  PubMed  Google Scholar 

  25. Wightman B, Ha I, Ruvkun G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell. 1993;75:855–62.

    Article  CAS  PubMed  Google Scholar 

  26. Kim VN, Han J, Siomi MC. Biogenesis of small RNAs in animals. Nat Rev Mol Cell Biol. 2009;10:126–39. https://doi.org/10.1038/nrm2632.

    Article  CAS  PubMed  Google Scholar 

  27. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–9. https://doi.org/10.1038/ncb1596.

    Article  CAS  PubMed  Google Scholar 

  28. Godlewski J, Krichevsky AM, Johnson MD, Chiocca EA, Bronisz A. Belonging to a network–microRNAs, extracellular vesicles, and the glioblastoma microenvironment. Neuro Oncol. 2015;17:652–62. https://doi.org/10.1093/neuonc/nou292.

    Article  CAS  PubMed  Google Scholar 

  29. He L, Thomson JM, Hemann MT, Hernando-Monge E, Mu D, Goodson S, Powers S, Cordon-Cardo C, Lowe SW, Hannon GJ, Hammond SM. A microRNA polycistron as a potential human oncogene. Nature. 2005;435:828–33. https://doi.org/10.1038/nature03552.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ghaemmaghami AB, Mahjoubin-Tehran M, Movahedpour A, Morshedi K, Sheida A, Taghavi SP, Mirzaei H, Hamblin MR. Role of exosomes in malignant glioma: microRNAs and proteins in pathogenesis and diagnosis. Cell Commun Signal. 2020;18:120. https://doi.org/10.1186/s12964-020-00623-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Godnic I, Zorc M, Jevsinek Skok D, Calin GA, Horvat S, Dovc P, Kovac M, Kunej T. Genome-wide and species-wide in silico screening for intragenic MicroRNAs in human, mouse and chicken. PLoS ONE. 2013;8:e65165. https://doi.org/10.1371/journal.pone.0065165.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Simion V, Henriet E, Juric V, Aquino R, Loussouarn C, Laurent Y, Martin F, Midoux P, Garcion E, Pichon C, Baril P. Intracellular trafficking and functional monitoring of miRNA delivery in glioblastoma using lipopolyplexes and the miRNA-ON RILES reporter system. J Control Release. 2020;327:429–43. https://doi.org/10.1016/j.jconrel.2020.08.028.

    Article  CAS  PubMed  Google Scholar 

  33. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–97.

    Article  CAS  PubMed  Google Scholar 

  34. Bronisz A, Wang Y, Nowicki MO, Peruzzi P, Ansari K, Ogawa D, Balaj L, De Rienzo G, Mineo M, Nakano I, Ostrowski MC, Hochberg F, Weissleder R, Lawler SE, Chiocca EA, Godlewski J. Extracellular vesicles modulate the glioblastoma microenvironment via a tumor suppression signaling network directed by miR-1. Cancer Res. 2014;74:738–50. https://doi.org/10.1158/0008-5472.CAN-13-2650.

    Article  CAS  PubMed  Google Scholar 

  35. Calin GA, Dumitru CD, Shimizu M, et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA. 2002;99:15524–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Thakur BK, Zhang H, Becker A, Matei I, Huang Y, Costa-Silva B, Zheng Y, Hoshino A, Brazier H, Xiang J, Williams C, Rodriguez-Barrueco R, Silva JM, Zhang W, Hearn S, Elemento O, Paknejad N, Manova-Todorova K, Welte K, Bromberg J, Peinado H, Lyden D. Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell Res. 2014;24:766–9. https://doi.org/10.1038/cr.2014.44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Panzarini E, Tacconi S, Carata E, Mariano S, Tata AM, Dini L. Molecular characterization of temozolomide-treated and non temozolomide-treated glioblastoma cells released extracellular vesicles and their role in the macrophage response. Int J Mol Sci. 2020;21(21):8353. https://doi.org/10.3390/ijms21218353.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhang L, Yu D. Exosomes in cancer development, metastasis, and immunity. Biochim Biophys Acta Rev Cancer. 1871;2019:455–68. https://doi.org/10.1016/j.bbcan.2019.04.004.

    Article  CAS  Google Scholar 

  39. Chistiakov DA, Chekhonin VP. Extracellular vesicles shed by glioma cells: pathogenic role and clinical value. Tumour Biol. 2014;35:8425–38. https://doi.org/10.1007/s13277-014-2262-9.

    Article  CAS  PubMed  Google Scholar 

  40. Santangelo A, Imbruce P, Gardenghi B, Belli L, Agushi R, Tamanini A, Munari S, Bossi AM, Scambi I, Benati D, Mariotti R, Di Gennaro G, Sbarbati A, Eccher A, Ricciardi GK, Ciceri EM, Sala F, Pinna G, Lippi G, Cabrini G, Dechecchi MC. A microRNA signature from serum exosomes of patients with glioma as complementary diagnostic biomarker. J Neurooncol. 2018;136:51–62. https://doi.org/10.1007/s11060-017-2639-x.

    Article  CAS  PubMed  Google Scholar 

  41. Skog J, Wurdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, Curry WT Jr, Carter BS, Krichevsky AM, Breakefield XO. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10:1470–6. https://doi.org/10.1038/ncb1800.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Saadatpour L, Fadaee E, Fadaei S, Nassiri Mansour R, Mohammadi M, Mousavi SM, Goodarzi M, Verdi J, Mirzaei H. Glioblastoma: exosome and microRNA as novel diagnosis biomarkers. Cancer Gene Ther. 2016;23:415–8. https://doi.org/10.1038/cgt.2016.48.

    Article  CAS  PubMed  Google Scholar 

  43. Li Z, Ye L, Wang L, Quan R, Zhou Y, Li X. Identification of miRNA signatures in serum exosomes as a potential biomarker after radiotherapy treatment in glioma patients. Ann Diagn Pathol. 2020;44: 151436. https://doi.org/10.1016/j.anndiagpath.2019.151436.

    Article  PubMed  Google Scholar 

  44. Ha M, Kim VN. Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol. 2014;15:509–24. https://doi.org/10.1038/nrm3838.

    Article  CAS  PubMed  Google Scholar 

  45. Rupaimoole R, Slack FJ. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 2017;16:203–22. https://doi.org/10.1038/nrd.2016.246.

    Article  CAS  PubMed  Google Scholar 

  46. Freidlin B, Korn EL. Biomarker enrichment strategies: matching trial design to biomarker credentials. Nat Rev Clin Oncol. 2014;11:81–90. https://doi.org/10.1038/nrclinonc.2013.218.

    Article  PubMed  Google Scholar 

  47. Hu P, Wang K, Zhou D, Wang L, Zhao M, Wang W, Zhang Y, Liu Y, Yu R, Zhou X. GOLPH3 regulates exosome miRNA secretion in glioma cells. J Mol Neurosci. 2020;70:1257–66. https://doi.org/10.1007/s12031-020-01535-6.

    Article  CAS  PubMed  Google Scholar 

  48. Caponnetto F, Dalla E, Mangoni D, Piazza S, Radovic S, Ius T, Skrap M, Di Loreto C, Beltrami AP, Manini I, Cesselli D. The miRNA content of exosomes released from the glioma microenvironment can affect malignant progression. Biomedicines. 2020;8(12):564. https://doi.org/10.3390/biomedicines8120564.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Godlewski J, Ferrer-Luna R, Rooj AK, Mineo M, Ricklefs F, Takeda YS, Nowicki MO, Salinska E, Nakano I, Lee H, Weissleder R, Beroukhim R, Chiocca EA, Bronisz A. MicroRNA signatures and molecular subtypes of glioblastoma: the role of extracellular transfer. Stem Cell Reports. 2017;8:1497–505. https://doi.org/10.1016/j.stemcr.2017.04.024.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Li X, Ling N, Bai Y, Dong W, Hui GZ, Liu D, Zhao J, Hu J. MiR-16-1 plays a role in reducing migration and invasion of glioma cells. Anat Rec (Hoboken). 2013;296:427–32. https://doi.org/10.1002/ar.22626.

    Article  CAS  PubMed  Google Scholar 

  51. Lessi F, Aretini P, Rizzo M, Morelli M, Menicagli M, Franceschi S, Mazzanti CM. Analysis of exosome-derived microRNAs reveals insights of intercellular communication during invasion of breast, prostate and glioblastoma cancer cells. Cell Adh Migr. 2021;15:180–201. https://doi.org/10.1080/19336918.2021.1935407.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Li X, Guan J, Jiang Z, Cheng S, Hou W, Yao J, Wang Z. Microglial exosome miR-7239-3p promotes glioma progression by regulating circadian genes. Neurosci Bull. 2021;37:497–510. https://doi.org/10.1007/s12264-020-00626-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Jones J, Nguyen H, Drummond K, Morokoff A. Circulating biomarkers for glioma: a review. Neurosurgery. 2021;88:E221-e230. https://doi.org/10.1093/neuros/nyaa540.

    Article  PubMed  Google Scholar 

  54. Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, Downing JR, Jacks T, Horvitz HR, Golub TR. MicroRNA expression profiles classify human cancers. Nature. 2005;435:834–8. https://doi.org/10.1038/nature03702.

    Article  CAS  PubMed  Google Scholar 

  55. Wang S, Xu Z, Zhang C, Yu R, Jiang J, Wang C, Qu C. High-throughput sequencing-based identification of serum exosomal differential miRNAs in high-grade glioma and intracranial lymphoma. Biomed Res Int. 2020;2020:2102645. https://doi.org/10.1155/2020/2102645.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Dong L, Li Y, Han C, Wang X, She L, Zhang H. miRNA microarray reveals specific expression in the peripheral blood of glioblastoma patients. Int J Oncol. 2014;45:746–56. https://doi.org/10.3892/ijo.2014.2459.

    Article  CAS  PubMed  Google Scholar 

  57. Manterola L, Guruceaga E, Gallego Perez-Larraya J, Gonzalez-Huarriz M, Jauregui P, Tejada S, Diez-Valle R, Segura V, Sampron N, Barrena C, Ruiz I, Agirre A, Ayuso A, Rodriguez J, Gonzalez A, Xipell E, Matheu A, de Munain AL, Tunon T, Zazpe I, Garcia-Foncillas J, Paris S, Delattre JY, Alonso MM. A small noncoding RNA signature found in exosomes of GBM patient serum as a diagnostic tool. Neuro Oncol. 2014;16:520–7. https://doi.org/10.1093/neuonc/not218.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Akers JC, Ramakrishnan V, Kim R, Phillips S, Kaimal V, Mao Y, Hua W, Yang I, Fu CC, Nolan J, Nakano I, Yang Y, Beaulieu M, Carter BS, Chen CC. miRNA contents of cerebrospinal fluid extracellular vesicles in glioblastoma patients. J Neurooncol. 2015;123:205–16. https://doi.org/10.1007/s11060-015-1784-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Tumilson CA, Lea RW, Alder JE, Shaw L. Circulating microRNA biomarkers for glioma and predicting response to therapy. Mol Neurobiol. 2014;50:545–58. https://doi.org/10.1007/s12035-014-8679-8.

    Article  CAS  PubMed  Google Scholar 

  60. Masoudi MS, Mehrabian E, Mirzaei H. MiR-21: a key player in glioblastoma pathogenesis. J Cell Biochem. 2018;119:1285–90. https://doi.org/10.1002/jcb.26300.

    Article  CAS  PubMed  Google Scholar 

  61. Santangelo A, Imbrucè P, Gardenghi B, Belli L, Agushi R, Tamanini A, Munari S, Bossi AM, Scambi I, Benati D, Mariotti R, Di Gennaro G, Sbarbati A, Eccher A, Ricciardi GK, Ciceri EM, Sala F, Pinna G, Lippi G, Cabrini G, Dechecchi MC. A microRNA signature from serum exosomes of patients with glioma as complementary diagnostic biomarker. J Neurooncol. 2018;136:51–62. https://doi.org/10.1007/s11060-017-2639-x.

    Article  CAS  PubMed  Google Scholar 

  62. Akers JC, Ramakrishnan V, Kim R, Skog J, Nakano I, Pingle S, Kalinina J, Hua W, Kesari S, Mao Y, Breakefield XO, Hochberg FH, Van Meir EG, Carter BS, Chen CC. MiR-21 in the extracellular vesicles (EVs) of cerebrospinal fluid (CSF): a platform for glioblastoma biomarker development. PLoS ONE. 2013;8: e78115. https://doi.org/10.1371/journal.pone.0078115.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Gabriely G, Yi M, Narayan RS, Niers JM, Wurdinger T, Imitola J, Ligon KL, Kesari S, Esau C, Stephens RM, Tannous BA, Krichevsky AM. Human glioma growth is controlled by microRNA-10b. Cancer Res. 2011;71:3563–72. https://doi.org/10.1158/0008-5472.CAN-10-3568.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Tabibkhooei A, Izadpanahi M, Arab A, Zare-Mirzaei A, Minaeian S, Rostami A, Mohsenian A. Profiling of novel circulating microRNAs as a non-invasive biomarker in diagnosis and follow-up of high and low-grade gliomas. Clin Neurol Neurosurg. 2020;190: 105652. https://doi.org/10.1016/j.clineuro.2019.105652.

    Article  PubMed  Google Scholar 

  65. Ciafre SA, Galardi S, Mangiola A, Ferracin M, Liu CG, Sabatino G, Negrini M, Maira G, Croce CM, Farace MG. Extensive modulation of a set of microRNAs in primary glioblastoma. Biochem Biophys Res Commun. 2005;334:1351–8. https://doi.org/10.1016/j.bbrc.2005.07.030.

    Article  CAS  PubMed  Google Scholar 

  66. Yang JK, Yang JP, Tong J, Jing SY, Fan B, Wang F, Sun GZ, Jiao BH. Exosomal miR-221 targets DNM3 to induce tumor progression and temozolomide resistance in glioma. J Neurooncol. 2017;131:255–65. https://doi.org/10.1007/s11060-016-2308-5.

    Article  CAS  PubMed  Google Scholar 

  67. Wortzel I, Dror S, Kenific CM, Lyden D. Exosome-mediated metastasis: communication from a distance. Dev Cell. 2019;49:347–60. https://doi.org/10.1016/j.devcel.2019.04.011.

    Article  CAS  PubMed  Google Scholar 

  68. Cai Q, Zhu A, Gong L. Exosomes of glioma cells deliver miR-148a to promote proliferation and metastasis of glioblastoma via targeting CADM1. Bull Cancer. 2018;105:643–51. https://doi.org/10.1016/j.bulcan.2018.05.003.

    Article  PubMed  Google Scholar 

  69. Kim J, Zhang Y, Skalski M, Hayes J, Kefas B, Schiff D, Purow B, Parsons S, Lawler S, Abounader R. microRNA-148a is a prognostic oncomiR that targets MIG6 and BIM to regulate EGFR and apoptosis in glioblastoma. Cancer Res. 2014;74:1541–53. https://doi.org/10.1158/0008-5472.CAN-13-1449.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Teplyuk NM, Mollenhauer B, Gabriely G, Giese A, Kim E, Smolsky M, Kim RY, Saria MG, Pastorino S, Kesari S, Krichevsky AM. MicroRNAs in cerebrospinal fluid identify glioblastoma and metastatic brain cancers and reflect disease activity. Neuro Oncol. 2012;14:689–700. https://doi.org/10.1093/neuonc/nos074.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Nikolova E, Georgiev C, Laleva L, Milev M, Spiriev T, Stoyanov S, Taseva-Mineva T, Mitev V, Todorova A. Diagnostic, grading and prognostic role of a restricted miRNAs signature in primary and metastatic brain tumours. Discussion on their therapeutic perspectives. Mol Genet Genomics. 2022;297:357–71. https://doi.org/10.1007/s00438-021-01851-5.

    Article  CAS  PubMed  Google Scholar 

  72. Bao Z, Zhang N, Niu W, Mu M, Zhang X, Hu S, Niu C. Exosomal miR-155-5p derived from glioma stem-like cells promotes mesenchymal transition via targeting ACOT12. Cell Death Dis. 2022;13:725. https://doi.org/10.1038/s41419-022-05097-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Hu X, Martinez-Ledesma E, Zheng S, Kim H, Barthel F, Jiang T, Hess KR, Verhaak RGW. Multigene signature for predicting prognosis of patients with 1p19q co-deletion diffuse glioma. Neuro Oncol. 2017;19:786–95. https://doi.org/10.1093/neuonc/now285.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. He X, Qi Y, Zhang X, Liu X, Li X, Li S, Wu Y, Zhang Q. Current landscape of tumor-derived exosomal ncRNAs in glioma progression, detection, and drug resistance. Cell Death Dis. 2021;12:1145. https://doi.org/10.1038/s41419-021-04430-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Karachi A, Dastmalchi F, Mitchell DA, Rahman M. Temozolomide for immunomodulation in the treatment of glioblastoma. Neuro Oncol. 2018;20:1566–72. https://doi.org/10.1093/neuonc/noy072.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Yang Z, Shi J, Xie J, Wang Y, Sun J, Liu T, Zhao Y, Zhao X, Wang X, Ma Y, Malkoc V, Chiang C, Deng W, Chen Y, Fu Y, Kwak KJ, Fan Y, Kang C, Yin C, Rhee J, Bertani P, Otero J, Lu W, Yun K, Lee AS, Jiang W, Teng L, Kim BYS, Lee LJ. Large-scale generation of functional mRNA-encapsulating exosomes via cellular nanoporation. Nat Biomed Eng. 2020;4:69–83. https://doi.org/10.1038/s41551-019-0485-1.

    Article  CAS  PubMed  Google Scholar 

  77. Kamerkar S, LeBleu VS, Sugimoto H, Yang S, Ruivo CF, Melo SA, Lee JJ, Kalluri R. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature. 2017;546:498–503. https://doi.org/10.1038/nature22341.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Sana J, Hajduch M, Michalek J, Vyzula R, Slaby O. MicroRNAs and glioblastoma: roles in core signalling pathways and potential clinical implications. J Cell Mol Med. 2011;15:1636–44. https://doi.org/10.1111/j.1582-4934.2011.01317.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Guessous F, Zhang Y, Kofman A, Catania A, Li Y, Schiff D, Purow B, Abounader R. microRNA-34a is tumor suppressive in brain tumors and glioma stem cells. Cell Cycle. 2010;9:1031–6. https://doi.org/10.4161/cc.9.6.10987.

    Article  CAS  PubMed  Google Scholar 

  80. Silber J, Jacobsen A, Ozawa T, Harinath G, Pedraza A, Sander C, Holland EC, Huse JT. miR-34a repression in proneural malignant gliomas upregulates expression of its target PDGFRA and promotes tumorigenesis. PLoS ONE. 2012;7: e33844. https://doi.org/10.1371/journal.pone.0033844.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Xu X, Liu Y, Li Y, Chen H, Zhang Y, Liu J, Deng S, Zheng Y, Sun X, Wang J, Chen T, Huang M, Ke Y. Selective exosome exclusion of miR-375 by glioma cells promotes glioma progression by activating the CTGF-EGFR pathway. J Exp Clin Cancer Res. 2021;40:16. https://doi.org/10.1186/s13046-020-01810-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Bouzari B. Angioregulatory role of miRNAs and exosomal miRNAs in glioblastoma pathogenesis. Biomed Pharmacother. 2022;148:0753–3322. https://doi.org/10.1016/j.biopha.2022.112760.

    Article  CAS  Google Scholar 

  83. Zhang G, Zhang Y, Cheng S, Wu Z, Liu F, Zhang J. CD133 positive U87 glioblastoma cells-derived exosomal microRNAs in hypoxia- versus normoxia-microenviroment. J Neurooncol. 2017;135:37–46. https://doi.org/10.1007/s11060-017-2566-x.

    Article  CAS  PubMed  Google Scholar 

  84. Garofalo M, Croce CM. microRNAs: Master regulators as potential therapeutics in cancer. Annu Rev Pharmacol Toxicol. 2011;51:25–43. https://doi.org/10.1146/annurev-pharmtox-010510-100517.

    Article  CAS  PubMed  Google Scholar 

  85. Zhang Z, Yin J, Lu C, Wei Y, Zeng A, You Y. Exosomal transfer of long non-coding RNA SBF2-AS1 enhances chemoresistance to temozolomide in glioblastoma. J Exp Clin Cancer Res. 2019;38:166. https://doi.org/10.1186/s13046-019-1139-6.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Movahedpour A, Khatami SH, Khorsand M, Salehi M, Savardashtaki A, Mirmajidi SH, Negahdari B, Khanjani N, Naeli P, Vakili O, Taheri-Anganeh M. Exosomal noncoding RNAs: key players in glioblastoma drug resistance. Mol Cell Biochem. 2021;476:4081–92. https://doi.org/10.1007/s11010-021-04221-2.

    Article  CAS  PubMed  Google Scholar 

  87. Yin J, Zeng A, Zhang Z, Shi Z, Yan W, You Y. Exosomal transfer of miR-1238 contributes to temozolomide-resistance in glioblastoma. EBioMedicine. 2019;42:238–51. https://doi.org/10.1016/j.ebiom.2019.03.016.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Wang J, Li T, Wang B. Exosomal transfer of miR253p promotes the proliferation and temozolomide resistance of glioblastoma cells by targeting FBXW7. Int J Oncol. 2021;59(2):64. https://doi.org/10.3892/ijo.2021.5244.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Yu L, Gui S, Liu Y, et al. Exosomes derived from microRNA-199a-overexpressing mesenchymal stem cells inhibit glioma progression by down-regulating AGAP2. Aging. 2019;11(15):5300–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Munoz JL, Bliss SA, Greco SJ, Ramkissoon SH, Ligon KL, Rameshwar P. Delivery of functional anti-miR-9 by mesenchymal stem cell-derived exosomes to glioblastoma multiforme cells conferred chemosensitivity. Mol Ther Nucleic Acids. 2013;2: e126. https://doi.org/10.1038/mtna.2013.60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Sharif S, Ghahremani MH, Soleimani M. Delivery of exogenous miR-124 to glioblastoma multiform cells by Wharton’s jelly mesenchymal stem cells decreases cell proliferation and migration, and confers chemosensitivity. Stem Cell Rev Rep. 2018;14:236–46. https://doi.org/10.1007/s12015-017-9788-3.

    Article  CAS  PubMed  Google Scholar 

  92. Yan T, Wu M, Lv S, et al. Exosomes derived from microRNA-512–5p-transfected bone mesenchymal stem cells inhibit glioblastoma progression by targeting JAG1. Aging. 2021;13:9911–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Figueroa J, Phillips LM, Shahar T, Hossain A, Gumin J, Kim H, Bean AJ, Calin GA, Fueyo J, Walters ET, Kalluri R, Verhaak RG, Lang FF. Exosomes from glioma-associated mesenchymal stem cells increase the tumorigenicity of glioma stem-like cells via transfer of miR-1587. Cancer Res. 2017;77:5808–19. https://doi.org/10.1158/0008-5472.CAN-16-2524.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Wei J, Wang F, Kong LY, Xu S, Doucette T, Ferguson SD, Yang Y, McEnery K, Jethwa K, Gjyshi O, Qiao W, Levine NB, Lang FF, Rao G, Fuller GN, Calin GA, Heimberger AB. miR-124 inhibits STAT3 signaling to enhance T cell-mediated immune clearance of glioma. Cancer Res. 2013;73:3913–26. https://doi.org/10.1158/0008-5472.CAN-12-4318.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Lang FM, Hossain A, Gumin J, Momin EN, Shimizu Y, Ledbetter D, Shahar T, Yamashita S, Parker Kerrigan B, Fueyo J, Sawaya R, Lang FF. Mesenchymal stem cells as natural biofactories for exosomes carrying miR-124a in the treatment of gliomas. Neuro Oncol. 2018;20:380–90. https://doi.org/10.1093/neuonc/nox152.

    Article  CAS  PubMed  Google Scholar 

  96. Sun X, Ma X, Wang J, et al. Glioma stem cells-derived exosomes promote the angiogenic ability of endothelial cells through miR-21/VEGF signal. Oncotarget. 2017;8:36137–48.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Jiang J, Lu J, Wang X, et al. Glioma stem cell-derived exosomal miR-944 reduces glioma growth and angiogenesis by inhibiting AKT/ERK signaling. Aging. 2021;13(15):19243–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Zhang Z, Guo X, Guo X, et al. MicroRNA-29a-3p delivery via exosomes derived from engineered human mesenchymal stem cells exerts tumour suppressive effects by inhibiting migration and vasculogenic mimicry in glioma. Aging. 2021;13(4):5055–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Kim R, Lee S, Lee J, Kim M, Kim WJ, Lee HW, Lee MY, Kim J, Chang W. Exosomes derived from microRNA-584 transfected mesenchymal stem cells: novel alternative therapeutic vehicles for cancer therapy. BMB Rep. 2018;51:406–11. https://doi.org/10.5483/bmbrep.2018.51.8.105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Bao L, Li X. MicroRNA-32 targeting PTEN enhances M2 macrophage polarization in the glioma microenvironment and further promotes the progression of glioma. Mol Cell Biochem. 2019;460:67–79. https://doi.org/10.1007/s11010-019-03571-2.

    Article  CAS  PubMed  Google Scholar 

  101. Zhu C, Mustafa D, Zheng PP, van der Weiden M, Sacchetti A, Brandt M, Chrifi I, Tempel D, Leenen PJM, Duncker DJ, Cheng C, Kros JM. Activation of CECR1 in M2-like TAMs promotes paracrine stimulation-mediated glial tumor progression. Neuro Oncol. 2017;19:648–59. https://doi.org/10.1093/neuonc/now251.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Yao J, Wang Z, Cheng Y, Ma C, Zhong Y, Xiao Y, Gao X, Li Z. M2 macrophage-derived exosomal microRNAs inhibit cell migration and invasion in gliomas through PI3K/AKT/mTOR signaling pathway. J Transl Med. 2021;19:99. https://doi.org/10.1186/s12967-021-02766-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Qi Y, Jin C, Qiu W, Zhao R, Wang S, Li B, Zhang Z, Guo Q, Zhang S, Gao Z, Zhao S, Pan Z, Fan Y, Chen Z, Wang H, Xu J, Deng L, Ni S, Wang J, Xue H, Xue F, Li G. The dual role of glioma exosomal microRNAs: glioma eliminates tumor suppressor miR-1298-5p via exosomes to promote immunosuppressive effects of MDSCs. Cell Death Dis. 2022;13:426. https://doi.org/10.1038/s41419-022-04872-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Guo X, Qiu W, Liu Q, Qian M, Wang S, Zhang Z, Gao X, Chen Z, Xue H, Li G. Immunosuppressive effects of hypoxia-induced glioma exosomes through myeloid-derived suppressor cells via the miR-10a/Rora and miR-21/Pten pathways. Oncogene. 2018;37:4239–59. https://doi.org/10.1038/s41388-018-0261-9.

    Article  CAS  PubMed  Google Scholar 

  105. Guo X, Qiu W, Wang J, Liu Q, Qian M, Wang S, Zhang Z, Gao X, Chen Z, Guo Q, Xu J, Xue H, Li G. Glioma exosomes mediate the expansion and function of myeloid-derived suppressor cells through microRNA-29a/Hbp1 and microRNA-92a/Prkar1a pathways. Int J Cancer. 2019;144:3111–26. https://doi.org/10.1002/ijc.32052.

    Article  CAS  PubMed  Google Scholar 

  106. Yue X, Lan F, Xia T. Hypoxic glioma cell-secreted exosomal miR-301a activates Wnt/beta-catenin signaling and promotes radiation resistance by targeting TCEAL7. Mol Ther. 2019;27:1939–49. https://doi.org/10.1016/j.ymthe.2019.07.011.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Qian M, Chen Z, Guo X, Wang S, Zhang Z, Qiu W, Qi Y, Zhang S, Xu J, Zhao R, Xue H, Li G. Exosomes derived from hypoxic glioma deliver miR-1246 and miR-10b-5p to normoxic glioma cells to promote migration and invasion. Lab Invest. 2021;101:612–24. https://doi.org/10.1038/s41374-020-00522-0.

    Article  CAS  PubMed  Google Scholar 

  108. Shi R, Wang P-Y, Li X-Y, et al. Exosomal levels of miRNA-21 from cerebrospinal fluids associated with poor prognosis and tumor recurrence of glioma patients. Oncotarget. 2015;6(29):26971–81.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Lan F, Qing Q, Pan Q, Hu M, Yu H, Yue X. Serum exosomal miR-301a as a potential diagnostic and prognostic biomarker for human glioma. Cell Oncol (Dordr). 2018;41:25–33. https://doi.org/10.1007/s13402-017-0355-3.

    Article  CAS  PubMed  Google Scholar 

  110. Stakaitis R, Pranckeviciene A, Steponaitis G, Tamasauskas A, Bunevicius A, Vaitkiene P. Unique Interplay between molecular miR-181b/d biomarkers and health related quality of life score in the predictive glioma models. Int J Mol Sci. 2020;21(20):7450. https://doi.org/10.3390/ijms21207450.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Henriksen M, Johnsen KB, Olesen P, Pilgaard L, Duroux M. MicroRNA expression signatures and their correlation with clinicopathological features in glioblastoma multiforme. Neuromolecular Med. 2014;16:565–77. https://doi.org/10.1007/s12017-014-8309-7.

    Article  CAS  PubMed  Google Scholar 

  112. Li J, Yuan H, Xu H, Zhao H, Xiong N. Hypoxic cancer-secreted exosomal miR-182-5p Promotes glioblastoma angiogenesis by targeting Kruppel-like factor 2 and 4. Mol Cancer Res. 2020;18:1218–31. https://doi.org/10.1158/1541-7786.MCR-19-0725.

    Article  CAS  PubMed  Google Scholar 

  113. Shao N, Xue L, Wang R, Luo K, Zhi F, Lan Q. miR-454-3p is an exosomal biomarker and functions as a tumor suppressor in glioma. Mol Cancer Ther. 2019;18:459–69. https://doi.org/10.1158/1535-7163.MCT-18-0725.

    Article  CAS  PubMed  Google Scholar 

  114. Garcia CM, Toms SA. The role of circulating microRNA in glioblastoma liquid biopsy. World Neurosurg. 2020;138:425–35. https://doi.org/10.1016/j.wneu.2020.03.128.

    Article  PubMed  Google Scholar 

  115. Guessous F, Alvarado-Velez M, Marcinkiewicz L, Zhang Y, Kim J, Heister S, Kefas B, Godlewski J, Schiff D, Purow B, Abounader R. Oncogenic effects of miR-10b in glioblastoma stem cells. J Neurooncol. 2013;112:153–63. https://doi.org/10.1007/s11060-013-1047-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Zottel A, Samec N, Kump A, Dall’Olio LRR, Dominkus PP, Romih R, Hudoklin S, Mlakar J, Nikitin D, Sorokin M, Buzdin A, Jovcevska I, Komel R. Analysis of miR-9-5p, miR-124-3p, miR-21-5p, miR-138-5p, and miR-1-3p in glioblastoma cell lines and extracellular vesicles. Int J Mol Sci. 2020;21(22):8491. https://doi.org/10.3390/ijms21228491.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Sun J, Sun Z, Gareev I, Yan T, Chen X, Ahmad A, Zhang D, Zhao B, Beylerli O, Yang G, Zhao S. Exosomal miR-2276-5p in plasma is a potential diagnostic and prognostic biomarker in glioma. Front Cell Dev Biol. 2021;9: 671202. https://doi.org/10.3389/fcell.2021.671202.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Qiu S, Lin S, Hu D, Feng Y, Tan Y, Peng Y. Interactions of miR-323/miR-326/miR-329 and miR-130a/miR-155/miR-210 as prognostic indicators for clinical outcome of glioblastoma patients. J Transl Med. 2013;11:10. https://doi.org/10.1186/1479-5876-11-10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. van den Bent MJ, Geurts M, French PJ, Smits M, Capper D, Bromberg JEC, Chang SM. Primary brain tumours in adults. Lancet. 2023;402:1564–79. https://doi.org/10.1016/S0140-6736(23)01054-1.

    Article  PubMed  Google Scholar 

  120. McDonald MF, Hossain A, Momin EN, Hasan I, Singh S, Adachi S, Gumin J, Ledbetter D, Yang J, Long L, Daou M, Gopakumar S, Phillips LM, Kerrigan BP, Lang FF. Tumor-specific polycistronic miRNA delivered by engineered exosomes for the treatment of glioblastoma. Neuro Oncol. 2024;26:236–50. https://doi.org/10.1093/neuonc/noad199.

    Article  PubMed  Google Scholar 

  121. Peng L, Yuan XQ, Li GC. The emerging landscape of circular RNA ciRS-7 in cancer (review). Oncol Rep. 2015;33:2669–74. https://doi.org/10.3892/or.2015.3904.

    Article  CAS  PubMed  Google Scholar 

  122. Yoon S, Nguyen HCT, Jo W, Kim J, Chi SM, Park J, Kim SY, Nam D. Biclustering analysis of transcriptome big data identifies condition-specific microRNA targets. Nucleic Acids Res. 2019;47: e53. https://doi.org/10.1093/nar/gkz139.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Nicot C. RNA-Seq reveal the circular RNAs landscape of lung cancer. Mol Cancer. 2019;18:183. https://doi.org/10.1186/s12943-019-1118-8.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Vo JN, Cieslik M, Zhang Y, Shukla S, Xiao L, Zhang Y, Wu YM, Dhanasekaran SM, Engelke CG, Cao X, Robinson DR, Nesvizhskii AI, Chinnaiyan AM. The landscape of circular RNA in cancer. Cell. 2019;176:869–81. https://doi.org/10.1016/j.cell.2018.12.021.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Van Der Steen N, Lyu Y, Hitzler AK, Becker AC, Seiler J, Diederichs S. The circular RNA landscape of non-small cell lung cancer cells. Cancers (Basel). 2020;12(5):1091. https://doi.org/10.3390/cancers12051091.

    Article  CAS  PubMed  Google Scholar 

  126. Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2:569–79. https://doi.org/10.1038/nri855.

    Article  CAS  PubMed  Google Scholar 

  127. Syn NL, Wang L, Chow EK, Lim CT, Goh BC. Exosomes in cancer nanomedicine and immunotherapy: prospects and challenges. Trends Biotechnol. 2017;35:665–76. https://doi.org/10.1016/j.tibtech.2017.03.004.

    Article  CAS  PubMed  Google Scholar 

  128. Yang P, Lei H, Fu Y, Chen C, Tang L, Xia S, Guo Y, Chen G, Xie M, Yang J, Li F, Li L. Exosomal miR-151-3p in saliva: a potential non-invasive marker for gastric cancer diagnosis and prognosis modulated by Sijunzi decoction (SJZD) in mice. Heliyon. 2024;10: e29169. https://doi.org/10.1016/j.heliyon.2024.e29169.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Wang S, Ma F, Feng Y, Liu T, He S. Role of exosomal miR-21 in the tumor microenvironment and osteosarcoma tumorigenesis and progression (Review). Int J Oncol. 2020;56:1055–63. https://doi.org/10.3892/ijo.2020.4992.

    Article  CAS  PubMed  Google Scholar 

  130. Zhang Y, Liu Q, Zhang X, Huang H, Tang S, Chai Y, Xu Z, Li M, Chen X, Liu J, Yang C. Recent advances in exosome-mediated nucleic acid delivery for cancer therapy. J Nanobiotechnol. 2022;20:279. https://doi.org/10.1186/s12951-022-01472-z.

    Article  CAS  Google Scholar 

  131. Kefas B, Godlewski J, Comeau L, Li Y, Abounader R, Hawkinson M, Lee J, Fine H, Chiocca EA, Lawler S, Purow B. microRNA-7 inhibits the epidermal growth factor receptor and the Akt pathway and is down-regulated in glioblastoma. Cancer Res. 2008;68:3566–72. https://doi.org/10.1158/0008-5472.CAN-07-6639.

    Article  CAS  PubMed  Google Scholar 

  132. Zhang X, Zhang X, Hu S, Zheng M, Zhang J, Zhao J, Zhang X, Yan B, Jia L, Zhao J, Wu K, Yang A, Zhang R. Identification of miRNA-7 by genome-wide analysis as a critical sensitizer for TRAIL-induced apoptosis in glioblastoma cells. Nucleic Acids Res. 2017;45:5930–44. https://doi.org/10.1093/nar/gkx317.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Xia H, Qi Y, Ng SS, Chen X, Chen S, Fang M, Li D, Zhao Y, Ge R, Li G, Chen Y, He ML, Kung HF, Lai L, Lin MC. MicroRNA-15b regulates cell cycle progression by targeting cyclins in glioma cells. Biochem Biophys Res Commun. 2009;380:205–10. https://doi.org/10.1016/j.bbrc.2008.12.169.

    Article  CAS  PubMed  Google Scholar 

  134. 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 Lett. 2013;329:146–54. https://doi.org/10.1016/j.canlet.2012.10.026.

    Article  CAS  PubMed  Google Scholar 

  135. Xiao S, Yang Z, Qiu X, et al. miR-29c contribute to glioma cells temozolomide sensitivity by targeting O6-methylguanine-DNA methyltransferases indirectly. Oncotarget. 2016;7(31):50229–38.

    Article  PubMed  PubMed Central  Google Scholar 

  136. Li Y, Guessous F, Zhang Y, Dipierro C, Kefas B, Johnson E, Marcinkiewicz L, Jiang J, Yang Y, Schmittgen TD, Lopes B, Schiff D, Purow B, Abounader R. MicroRNA-34a inhibits glioblastoma growth by targeting multiple oncogenes. Cancer Res. 2009;69:7569–76. https://doi.org/10.1158/0008-5472.CAN-09-0529.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Wang B, Wu ZH, Lou PY, Chai C, Han SY, Ning JF, Li M. Human bone marrow-derived mesenchymal stem cell-secreted exosomes overexpressing microRNA-34a ameliorate glioblastoma development via down-regulating MYCN. Cell Oncol (Dordr). 2019;42:783–99. https://doi.org/10.1007/s13402-019-00461-z.

    Article  CAS  PubMed  Google Scholar 

  138. Smits M, Nilsson J, Mir SE, et al. miR-101 is down-regulated in glioblastoma resulting in EZH2-induced proliferation, migration, and angiogenesis. Oncotarget. 2011;1(8):710–20.

    Article  PubMed Central  Google Scholar 

  139. Yang G, Zhang R, Chen X, Mu Y, Ai J, Shi C, Liu Y, Shi C, Sun L, Rainov NG, Li H, Yang B, Zhao S. MiR-106a inhibits glioma cell growth by targeting E2F1 independent of p53 status. J Mol Med (Berl). 2011;89:1037–50. https://doi.org/10.1007/s00109-011-0775-x.

    Article  CAS  PubMed  Google Scholar 

  140. Fowler A, Thomson D, Giles K, Maleki S, Mreich E, Wheeler H, Leedman P, Biggs M, Cook R, Little N, Robinson B, McDonald K. miR-124a is frequently down-regulated in glioblastoma and is involved in migration and invasion. Eur J Cancer. 2011;47:953–63. https://doi.org/10.1016/j.ejca.2010.11.026.

    Article  CAS  PubMed  Google Scholar 

  141. Wu N, Xiao L, Zhao X, Zhao J, Wang J, Wang F, Cao S, Lin X. miR-125b regulates the proliferation of glioblastoma stem cells by targeting E2F2. FEBS Lett. 2012;586:3831–9. https://doi.org/10.1016/j.febslet.2012.08.023.

    Article  CAS  PubMed  Google Scholar 

  142. Papagiannakopoulos T, Friedmann-Morvinski D, Neveu P, Dugas JC, Gill RM, Huillard E, Liu C, Zong H, Rowitch DH, Barres BA, Verma IM, Kosik KS. Pro-neural miR-128 is a glioma tumor suppressor that targets mitogenic kinases. Oncogene. 2012;31:1884–95. https://doi.org/10.1038/onc.2011.380.

    Article  CAS  PubMed  Google Scholar 

  143. Xu H, Zhao G, Zhang Y, Jiang H, Wang W, Zhao D, Hong J, Yu H, Qi L. Mesenchymal stem cell-derived exosomal microRNA-133b suppresses glioma progression via Wnt/beta-catenin signaling pathway by targeting EZH2. Stem Cell Res Ther. 2019;10:381. https://doi.org/10.1186/s13287-019-1446-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Yang Y, Wu J, Guan H, Cai J, Fang L, Li J, Li M. MiR-136 promotes apoptosis of glioma cells by targeting AEG-1 and Bcl-2. FEBS Lett. 2012;586:3608–12. https://doi.org/10.1016/j.febslet.2012.08.003.

    Article  CAS  PubMed  Google Scholar 

  145. Silber J, Lim DA, Petritsch C, Persson AI, Maunakea AK, Yu M, Vandenberg SR, Ginzinger DG, James CD, Costello JF, Bergers G, Weiss WA, Alvarez-Buylla A, Hodgson JG. miR-124 and miR-137 inhibit proliferation of glioblastoma multiforme cells and induce differentiation of brain tumor stem cells. BMC Med. 2008;6:14. https://doi.org/10.1186/1741-7015-6-14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Li RY, Chen LC, Zhang HY, Du WZ, Feng Y, Wang HB, Wen JQ, Liu X, Li XF, Sun Y, Yang DB, Jiang T, Li YL, Jiang CL. MiR-139 inhibits Mcl-1 expression and potentiates TMZ-induced apoptosis in glioma. CNS Neurosci Ther. 2013;19:477–83. https://doi.org/10.1111/cns.12089.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Wang L, Shi L-M, Jiang C-F. MiR-143 acts as a tumor suppressor by targeting N-RAS and enhances temozolomide-induced apoptosis in glioma. Oncotarget. 2014;5(14):5416–27.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Katakowski M, Buller B, Zheng X, Lu Y, Rogers T, Osobamiro O, Shu W, Jiang F, Chopp M. Exosomes from marrow stromal cells expressing miR-146b inhibit glioma growth. Cancer Lett. 2013;335:201–4. https://doi.org/10.1016/j.canlet.2013.02.019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Fareh M, Turchi L, Virolle V, Debruyne D, Almairac F, de la Forest Divonne S, Paquis P, Preynat-Seauve O, Krause KH, Chneiweiss H, Virolle T. The miR 302–367 cluster drastically affects self-renewal and infiltration properties of glioma-initiating cells through CXCR4 repression and consequent disruption of the SHH-GLI-NANOG network. Cell Death Differ. 2012;19(2):232–44. https://doi.org/10.1038/cdd.2011.89.

    Article  CAS  PubMed  Google Scholar 

  150. Fareh M, Almairac F, Turchi L, Burel-Vandenbos F, Paquis P, Fontaine D, Lacas-Gervais S, Junier MP, Chneiweiss H, Virolle T. Cell-based therapy using miR-302-367 expressing cells represses glioblastoma growth. Cell Death Dis. 2017;8: e2713. https://doi.org/10.1038/cddis.2017.117.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Chen L, Zhang J, Feng Y, Li R, Sun X, Du W, Piao X, Wang H, Yang D, Sun Y, Li X, Jiang T, Kang C, Li Y, Jiang C. MiR-410 regulates MET to influence the proliferation and invasion of glioma. Int J Biochem Cell Biol. 2012;44:1711–7. https://doi.org/10.1016/j.biocel.2012.06.027.

    Article  CAS  PubMed  Google Scholar 

  152. Yin K, Liu X. CircMMP1 promotes the progression of glioma through miR-433/HMGB3 axis in vitro and in vivo. IUBMB Life. 2020;72:2508–24. https://doi.org/10.1002/iub.2383.

    Article  CAS  PubMed  Google Scholar 

  153. Gwinn DM, Shackelford DB, Egan DF, Mihaylova MM, Mery A, Vasquez DS, Turk BE, Shaw RJ. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol Cell. 2008;30:214–26. https://doi.org/10.1016/j.molcel.2008.03.003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Nan Y, Han L, Zhang A, Wang G, Jia Z, Yang Y, Yue X, Pu P, Zhong Y, Kang C. MiRNA-451 plays a role as tumor suppressor in human glioma cells. Brain Res. 2010;1359:14–21. https://doi.org/10.1016/j.brainres.2010.08.074.

    Article  CAS  PubMed  Google Scholar 

  155. van der Vos KE, Abels ER, Zhang X, Lai C, Carrizosa E, Oakley D, Prabhakar S, Mardini O, Crommentuijn MH, Skog J, Krichevsky AM, Stemmer-Rachamimov A, Mempel TR, El Khoury J, Hickman SE, Breakefield XO. Directly visualized glioblastoma-derived extracellular vesicles transfer RNA to microglia/macrophages in the brain. Neuro Oncol. 2016;18:58–69. https://doi.org/10.1093/neuonc/nov244.

    Article  CAS  PubMed  Google Scholar 

  156. Gal H, Pandi G, Kanner AA, Ram Z, Lithwick-Yanai G, Amariglio N, Rechavi G, Givol D. MIR-451 and Imatinib mesylate inhibit tumor growth of Glioblastoma stem cells. Biochem Biophys Res Commun. 2008;376:86–90. https://doi.org/10.1016/j.bbrc.2008.08.107.

    Article  CAS  PubMed  Google Scholar 

  157. Wang L, Shi M, Hou S, Ding B, Liu L, Ji X, Zhang J, Deng Y. MiR-483-5p suppresses the proliferation of glioma cells via directly targeting ERK1. FEBS Lett. 2012;586:1312–7. https://doi.org/10.1016/j.febslet.2012.03.035.

    Article  CAS  PubMed  Google Scholar 

  158. Li X, Liu Y, Granberg KJ, Wang Q, Moore LM, Ji P, Gumin J, Sulman EP, Calin GA, Haapasalo H, Nykter M, Shmulevich I, Fuller GN, Lang FF, Zhang W. Two mature products of MIR-491 coordinate to suppress key cancer hallmarks in glioblastoma. Oncogene. 2015;34:1619–28. https://doi.org/10.1038/onc.2014.98.

    Article  CAS  PubMed  Google Scholar 

  159. Yan T, Wu M, Lv S, et al. Exosomes derived from microRNA-512–5p-transfected bone mesenchymal stem cells inhibit glioblastoma progression by targeting JAG1. Aging. 2021;13(7):9911–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Cunha PP, Costa PM, Morais CM, Lopes IR, Cardoso AM, Cardoso AL, Mano M, Jurado AS, Pedroso de Lima MC. High-throughput screening uncovers miRNAs enhancing glioblastoma cell susceptibility to tyrosine kinase inhibitors. Hum Mol Genet. 2017;26:4375–87. https://doi.org/10.1093/hmg/ddx323.

    Article  CAS  PubMed  Google Scholar 

  161. Ma W, Zhou Y, Liu M, Qin Q, Cui Y. Long non-coding RNA LINC00470 in serum derived exosome: a critical regulator for proliferation and autophagy in glioma cells. Cancer Cell Int. 2021;21:149. https://doi.org/10.1186/s12935-021-01825-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Wang X-P, Deng X-L, Li L-Y. MicroRNA-584 functions as a tumor suppressor and targets PTTG1IP in glioma. Int J Clin Exp Pathol. 2014;7(12):8573–82.

    PubMed  PubMed Central  Google Scholar 

  163. Chen X, Zhang Y, Shi Y, Lian H, Tu H, Han S, Peng B, Liu W, He X. MiR-873 acts as a novel sensitizer of glioma cells to cisplatin by targeting Bcl-2. Int J Oncol. 2015;47:1603–11. https://doi.org/10.3892/ijo.2015.3143.

    Article  CAS  PubMed  Google Scholar 

  164. Yan W, Zhang W, Sun L, Liu Y, You G, Wang Y, Kang C, You Y, Jiang T. Identification of MMP-9 specific microRNA expression profile as potential targets of anti-invasion therapy in glioblastoma multiforme. Brain Res. 2011;1411:108–15. https://doi.org/10.1016/j.brainres.2011.07.002.

    Article  CAS  PubMed  Google Scholar 

  165. Jiang J, Lu J, Wang X, et al. Glioma stem cell-derived exosomal miR-944 reduces glioma growth and angiogenesis by inhibiting AKT/ERK signaling. Aging. 2021;13:19243–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Chai Y, Wu HT, Liang CD, You CY, Xie MX, Xiao SW. Exosomal lncRNA ROR1-AS1 derived from tumor cells promotes glioma progression via regulating miR-4686. Int J Nanomedicine. 2020;15:8863–72. https://doi.org/10.2147/IJN.S271795.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Wang X, Cao Q, Shi Y, Wu X, Mi Y, Liu K, Kan Q, Fan R, Liu Z, Zhang M. Identification of low-dose radiation-induced exosomal circ-METRN and miR-4709-3p/GRB14/PDGFRalpha pathway as a key regulatory mechanism in Glioblastoma progression and radioresistance: functional validation and clinical theranostic significance. Int J Biol Sci. 2021;17:1061–78. https://doi.org/10.7150/ijbs.57168.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Chen X, Yang F, Zhang T, Wang W, Xi W, Li Y, Zhang D, Huo Y, Zhang J, Yang A, Wang T. MiR-9 promotes tumorigenesis and angiogenesis and is activated by MYC and OCT4 in human glioma. J Exp Clin Cancer Res. 2019;38:99. https://doi.org/10.1186/s13046-019-1078-2.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Tan X, Wang S, Yang B, Zhu L, Yin B, Chao T, Zhao J, Yuan J, Qiang B, Peng X. The CREB-miR-9 negative feedback minicircuitry coordinates the migration and proliferation of glioma cells. PLoS ONE. 2012;7: e49570. https://doi.org/10.1371/journal.pone.0049570.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Tan X, Wang S, Zhu L, Wu C, Yin B, Zhao J, Yuan J, Qiang B, Peng X. cAMP response element-binding protein promotes gliomagenesis by modulating the expression of oncogenic microRNA-23a. Proc Natl Acad Sci USA. 2012;109:15805–10. https://doi.org/10.1073/pnas.1207787109.

    Article  PubMed  PubMed Central  Google Scholar 

  171. Chen L, Han L, Zhang K, Shi Z, Zhang J, Zhang A, Wang Y, Song Y, Li Y, Jiang T, Pu P, Jiang C, Kang C. VHL regulates the effects of miR-23b on glioma survival and invasion via suppression of HIF-1alpha/VEGF and beta-catenin/Tcf-4 signaling. Neuro Oncol. 2012;14:1026–36. https://doi.org/10.1093/neuonc/nos122.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Chen L, Zhang A, Li Y, Zhang K, Han L, Du W, Yan W, Li R, Wang Y, Wang K, Pu P, Jiang T, Jiang C, Kang C. MiR-24 regulates the proliferation and invasion of glioma by ST7L via beta-catenin/Tcf-4 signaling. Cancer Lett. 2013;329:174–80. https://doi.org/10.1016/j.canlet.2012.10.025.

    Article  CAS  PubMed  Google Scholar 

  173. Wang ZF, Liao F, Wu H, Dai J. Glioma stem cells-derived exosomal miR-26a promotes angiogenesis of microvessel endothelial cells in glioma. J Exp Clin Cancer Res. 2019;38:201. https://doi.org/10.1186/s13046-019-1181-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Kim H, Huang W, Jiang X, Pennicooke B, Park PJ, Johnson MD. Integrative genome analysis reveals an oncomir/oncogene cluster regulating glioblastoma survivorship. Proc Natl Acad Sci USA. 2010;107:2183–8. https://doi.org/10.1073/pnas.0909896107.

    Article  PubMed  PubMed Central  Google Scholar 

  175. Huse JT, Brennan C, Hambardzumyan D, Wee B, Pena J, Rouhanifard SH, Sohn-Lee C, le Sage C, Agami R, Tuschl T, Holland EC. The PTEN-regulating microRNA miR-26a is amplified in high-grade glioma and facilitates gliomagenesis in vivo. Genes Dev. 2009;23:1327–37. https://doi.org/10.1101/gad.1777409.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Wang K, Wang X, Zou J, Zhang A, Wan Y, Pu P, Song Z, Qian C, Chen Y, Yang S, Wang Y. miR-92b controls glioma proliferation and invasion through regulating Wnt/beta-catenin signaling via Nemo-like kinase. Neuro Oncol. 2013;15:578–88. https://doi.org/10.1093/neuonc/not004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Koo S, Martin GS, Schulz KJ. Serial selection for invasiveness increases expression of miR-143/miR-145 in glioblastoma cell lines. BMC Cancer. 2012;12:143.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Wang M, Zhao Y, Yu ZY, Zhang RD, Li SA, Zhang P, Shan TK, Liu XY, Wang ZM, Zhao PC, Sun HW. Glioma exosomal microRNA-148a-3p promotes tumor angiogenesis through activating the EGFR/MAPK signaling pathway via inhibiting ERRFI1. Cancer Cell Int. 2020;20:518. https://doi.org/10.1186/s12935-020-01566-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Zeng A, Wei Z, Yan W, Yin J, Huang X, Zhou X, Li R, Shen F, Wu W, Wang X, You Y. Exosomal transfer of miR-151a enhances chemosensitivity to temozolomide in drug-resistant glioblastoma. Cancer Lett. 2018;436:10–21. https://doi.org/10.1016/j.canlet.2018.08.004.

    Article  CAS  PubMed  Google Scholar 

  180. Ebrahimkhani S, Vafaee F, Hallal S, Wei H, Lee MYT, Young PE, Satgunaseelan L, Beadnall H, Barnett MH, Shivalingam B, Suter CM, Buckland ME, Kaufman KL. Deep sequencing of circulating exosomal microRNA allows non-invasive glioblastoma diagnosis. NPJ Precis Oncol. 2018;2:28. https://doi.org/10.1038/s41698-018-0071-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Ujifuku K, Mitsutake N, Takakura S, Matsuse M, Saenko V, Suzuki K, Hayashi K, Matsuo T, Kamada K, Nagata I, Yamashita S. miR-195, miR-455-3p and miR-10a(*) are implicated in acquired temozolomide resistance in glioblastoma multiforme cells. Cancer Lett. 2010;296:241–8. https://doi.org/10.1016/j.canlet.2010.04.013.

    Article  CAS  PubMed  Google Scholar 

  182. Lan F, Yue X, Xia T. Exosomal microRNA-210 is a potentially non-invasive biomarker for the diagnosis and prognosis of glioma. Oncol Lett. 2020;19:1967–74. https://doi.org/10.3892/ol.2020.11249.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Yang JK, Liu HJ, Wang Y, Li C, Yang JP, Yang L, Qi XJ, Zhao YL, Shi XF, Li JC, Sun GZ, Jiao BH. Exosomal miR-214-5p released from glioblastoma cells modulates inflammatory response of microglia after lipopolysaccharide stimulation through targeting CXCR5. CNS Neurol Disord Drug Targets. 2019;18:78–87. https://doi.org/10.2174/1871527317666181105112009.

    Article  CAS  PubMed  Google Scholar 

  184. Olioso D, Caccese M, Santangelo A, Lippi G, Zagonel V, Cabrini G, Lombardi G, Dechecchi MC. Serum exosomal microRNA-21, 222 and 124–3p as noninvasive predictive biomarkers in newly diagnosed high-grade gliomas: a prospective study. Cancers (Basel). 2021;13(12):3006. https://doi.org/10.3390/cancers13123006.

    Article  CAS  PubMed  Google Scholar 

  185. Delic S, Lottmann N, Stelzl A, Liesenberg F, Wolter M, Gotze S, Zapatka M, Shiio Y, Sabel MC, Felsberg J, Reifenberger G, Riemenschneider MJ. MiR-328 promotes glioma cell invasion via SFRP1-dependent Wnt-signaling activation. Neuro Oncol. 2014;16:179–90. https://doi.org/10.1093/neuonc/not164.

    Article  CAS  PubMed  Google Scholar 

  186. Tang H, Liu X, Wang Z, She X, Zeng X, Deng M, Liao Q, Guo X, Wang R, Li X, Zeng F, Wu M, Li G. Interaction of hsa-miR-381 and glioma suppressor LRRC4 is involved in glioma growth. Brain Res. 2011;1390:21–32. https://doi.org/10.1016/j.brainres.2011.03.034.

    Article  CAS  PubMed  Google Scholar 

  187. Xue P, Huang S, Han X, Zhang C, Yang L, Xiao W, Fu J, Li H, Zhou Y. Exosomal miR-101-3p and miR-423-5p inhibit medulloblastoma tumorigenesis through targeting FOXP4 and EZH2. Cell Death Differ. 2022;29:82–95. https://doi.org/10.1038/s41418-021-00838-4.

    Article  CAS  PubMed  Google Scholar 

  188. Kushwaha D, Ramakrishnan V, Ng K, et al. A genome-wide miRNA screen revealed miR-603 as a MGMT regulating miRNA in glioblastoma. Oncotarget. 2014;5(12):4026–39.

    Article  PubMed  PubMed Central  Google Scholar 

  189. Jeansonne D, Pacifici M, Lassak A, Reiss K, Russo G, Zabaleta J, Peruzzi F. Differential effects of MicroRNAs on glioblastoma growth and migration. Genes (Basel). 2013;4:46–64. https://doi.org/10.3390/genes4010046.

    Article  CAS  PubMed  Google Scholar 

  190. Mrowczynski OD, Madhankumar AB, Sundstrom JM, et al. Exosomes impact survival to radiation exposure in cell line models of nervous system cancer. Oncotarget. 2018;9:36083–101.

    Article  PubMed  PubMed Central  Google Scholar 

  191. Thuringer D, Chanteloup G, Boucher J, et al. Modulation of the inwardly rectifying potassium channel Kir4.1 by the pro-invasive miR-5096 in glioblastoma cells. Oncotarget. 2017;8:37681–93.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (no. 82271395; 81401688; 8180051466 and 82001301); Science and Technology Program of Guangzhou (202105080053); Science and Technology Program of Guangzhou (202007030001); Guangdong Basic and Applied Basic Research Foundation(2023A1515030073); Youth Talent Two-way Exchange Project of Guangdong and Macao (KD0120230024) and the Special Project of Dengfeng Program of Guangdong Provincial People’s Hospital (KY0120220133, DFJHBF202111, KJ012020630).

Author information

Authors and Affiliations

Authors

Contributions

LY wrote the manuscript. LY and ZN analyzed the data and drafted the manuscript. ZXM and XJW analyzed the data. YF, GL and CTV reviewed and edited the manuscript. All authors participated in the conception of this manuscript and approved the final version of this manuscript.

Corresponding authors

Correspondence to Ge Li or Ying Feng.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, L., Niu, Z., Ma, Z. et al. Exploring the clinical implications and applications of exosomal miRNAs in gliomas: a comprehensive study. Cancer Cell Int 24, 323 (2024). https://doi.org/10.1186/s12935-024-03507-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-024-03507-x

Keywords