IMR Press / JIN / Volume 21 / Issue 4 / DOI: 10.31083/j.jin2104111
Open Access Review
Serum long non-coding RNAs as potential noninvasive biomarkers for glioblastoma diagnosis, prognosis, and chemoresistance
Show Less
1 Department of Neurological Surgery, University of Wisconsin, Madison, WI 53792, USA
*Correspondence: baskaya@neurosurgery.wisc.edu (Mustafa K. Baskaya)
Academic Editor: Rafael Franco
J. Integr. Neurosci. 2022, 21(4), 111; https://doi.org/10.31083/j.jin2104111
Submitted: 16 October 2021 | Revised: 28 January 2022 | Accepted: 31 January 2022 | Published: 7 June 2022
Copyright: © 2022 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Gliomas are common brain tumors with a variable prognosis based on their tumor grade. With glioblastomas, the prognosis is usually unfavorable. Thus, having accurate and rapid methods for their diagnosis and follow-up are essential for rapid discovery of the tumor and to protect patients from unnecessary procedures. Some glioma cases are challenging since there is a limited ability to differentiate between gliomas, recurrent glioblastomas, and single metastatic lesions. Monitoring treatment responses and follow-ups can also be challenging. While both radiological and serological markers have been identified that can aid diagnosis and assess therapies, a particularly promising new class of serological markers are long non-coding RNAs. Long non-coding RNAs are a relatively recently discovered class of regulatory RNA molecules that play critical roles in many cellular and physiological processes. The potential role that long non-coding RNAs play with glioma pathogenic processes is not fully understood. In this literature review, we highlight the potential for long non-coding RNAs to be used as serum biomarkers in glioblastoma patients, including their potential to serve as non-invasive, easy to use, and rapid diagnostic or prognostic indicators.

Keywords
non-coding RNA
lncRNA
glioblastoma
glioma
biomarker
microRNA
miRNA
1. Introduction

Gliomas originate from multiple glial cell types and can appear anywhere in the central nervous system [1]. They are the most common primary brain tumors in adults, comprising 80% of adult malignant brain tumors [1, 2]. Diffuse histological type glioblastomas (GBM) are the most aggressive and most fatal primary brain tumor. Fig. 1 shows a simplified algorithm for classification of diffuse gliomas according to the 2016 WHO Classification of Tumors of the Central Nervous System [3, 4]. There are two types of GBM, which are defined by the gene encoding for isocitrate dehydrogenase (IDH). These are: (1) IDH-wild types that represent >90% of cases, mainly affect the elderly, and rapidly develop de novo, which is why they are also called primary GBM, and (2) IDH-mutant types that arise in younger patients and represent about 10% of cases. IDH-mutant types develop through a progression from lower grade diffuse gliomas, hence why these are called secondary GBM [3, 4]. Surgical removal and radio-chemotherapy are the standard treatments for newly diagnosed GBMs [5]. Despite major advancements in radio-chemotherapy treatment, median survival has remained poor in the past few years with most tumors reoccurring [6, 7]. Many factors contribute to this treatment failure. These include the heterogeneity of the GBM microenvironment, development of resistance to available therapies, and the availability of intrinsic cancer stem cells with great regenerative potential [8].

Fig. 1.

A simplified algorithm for classification of diffuse gliomas based on histological features and for classification of GBM based on genetic features. Diffuse gliomas are classified histologically into astrocytoma, oligoastrocytoma, oligodendroglioma and glioblastoma. Glioblastomas are classified genetically into IDH wild type and IDH mutant. IDH refers to the gene that encodes isocitrate dehydrogenase.

Due to the severity, high recurrence rate, and poor prognosis of GBM, new and improved diagnostics are highly desired to enable early diagnosis and to monitor tumor progression, regression, and recurrence. Genetic biomarkers are a very promising diagnostic modality. Here we use the term biomarker for a substance that can objectively act as an indicator for a physiological or pathological process, such as a pharmacological response to a particular drug. Biomarkers may be found in tumor tissues or serum, and may include DNA, RNA, enzymes, metabolites, transcription factors, and cell surface receptors [9]. In the present work we will discuss serum long non-coding RNAs (lncRNAs) as promising biomarkers for the diagnosis and prognosis of GBM. We also will elucidate the pathophysiology by which those lncRNAs participate in carcinogenesis and anticancer drugs resistance.

1.1 Non-coding RNAs (ncRNA)

Only a small proportion of our genome is transcribed into mRNAs to produce protein. In contrast, most of our genome consists of ncRNAs that are not translated into proteins [10, 11]. In the past, there was a belief that the most important genetic material product were protein-encoding sequences, and that the remaining genome that does not encode proteins had unknown or obscure less important roles, such as ncRNAs and other gene products that were involved in gene expression and message transfer. But, more recently, it has been determined that ncRNA plays critical roles in diverse cellular and physiological processes including gene regulation, chromatin packaging, cell differentiation and development [10, 11, 12, 13, 14]. NcRNAs can be classified based on the number of nucleotides (nt) into lncRNAs that are greater than 200 nt long, and small ncRNAs that have no more than 200 nt. Small ncRNAs include microRNAs (miRNAs), small nuclear RNAs (snRNAs), small interfering RNAs (siRNAs), small nucleolar RNAs (snoRNAs), and Piwi-interacting RNAs (piRNAs) [15, 16, 17]. These different types of ncRNAs are illustrated in Fig. 2. In contrast to the short-lived miRNAs that have been extensively discussed in the literature, lncRNAs are stable and easily detectable in body fluids including plasma, and usually have higher tissue-specificity, thus these are a promising new class of biomarkers deserving of attention [18, 19, 20].

Fig. 2.

Categorization of types of non-coding RNAs (ncRNAs). NcRNAs are classified based on nucleotide number (nt) into long non-coding RNAs (lncRNAs) and small ncRNAs. Small ncRNAs are further classified based on their functions into many types: miRNAs (microRNAs), snRNAs (small nuclear RNAs), siRNAs (small interfering RNAs), snoRNAs (small nucleolar RNAs), piRNAs (Piwi-interacting RNAs).

1.2 Long noncoding RNAs

LncRNAs are non-protein coding RNA transcripts that are greater than 200 nt in length [21]. These play important roles in tumor proliferation and progression via regulating the expression of oncogenes, tumor suppressor genes, and other cancer-associated genes through epigenetic control, and through transcriptional, posttranscriptional, and translational regulation [22, 23]. Although most research has focused on the intracellular roles of lncRNAs, there is a growing interest in the diagnostic and prognostic role of circulating lncRNAs as potential non-invasive biomarkers for a variety of diseases and tumors. Specific lncRNAs have been shown to be survival predictors in patients with heart failure [24], mortality predictors in patients with acute kidney injury [25], and novel biomarkers for diagnosis of coronary artery disease [26], esophageal squamous cell carcinoma [27], and gastric cancer [28].

2. Long noncoding RNAs and glioblastomas

In gliomas, most of the reported lncRNAs can cause tumor progression or chemo-resistance, such as HOTAIR (HOX transcript antisense RNA), HOXA11-AS (HOXA11 antisense RNA), lncRNA NEAT1 (nuclear paraspeckle assembly transcript 1), lncRNA SBF2-AS1 (RNA SBF2 antisense RNA1), and lncRNA MALAT1 (metastasis-associated lung adenocarcinoma transcript) [29, 30, 31, 32, 33, 34]. In contrast to those lncRNAs, some lncRNAs have protective roles and their expression decrease may lead to tumor proliferation or chemo-resistance, such as lncRNA HERC2P2 [35].

A review of the literature indicates that there are several lncRNAs associated with GBM, but these levels were measured in biopsies or cell cultures. Using lncRNAs levels that are obtained from biopsies, while useful for research, is problematic for a clinical biomarker since obtaining a biopsy requires surgery, and the biopsy specimen itself, even without assaying for the lncRNA, is already sufficient for diagnosis. It thus would be of interest to determine if there are lncRNAs that might be used as reliable serum biomarkers for clinical diagnosis and prognosis, and as markers for chemoresistance.

2.1 HOX transcript antisense RNA (HOTAIR)

The HOTAIR gene is located within a HOXC gene cluster on chromosome 12 between the HOXC11 and HOXC12 genes [36, 37]. This gene encodes a long noncoding RNA (lncRNA) molecule called HOTAIR that stretches for nearly 2200 nucleotides [38]. As illustrated in Fig. 3, the HOTAIR-PRC2-LSD1 complex attaches to the promoters of many tumor suppressor genes enabling HOTAIR to silence those genes through methylation of H3K27me3 and demethylation of H3K4me2 [39, 40]. Because of HOTAIR’s role in carcinogenesis, HOTAIR has an important diagnostic or prognostic role in multiple tumors. For example, it acts as a potential biomarker to diagnose breast cancer [41]. It also correlates with disease progression in bladder cancer [42]. It is considered a novel diagnostic biomarker for esophageal squamous cell carcinoma [36], and generally, its aberrant expression is associated with the metastatic progression of many malignancies [43]. Regarding the level of HOTAIR expression in GBM tissues, in a study that used glioma samples to elucidate the correlation between HOTAIR and glioma, Zhang et al. [44] found a significant increase in HOTAIR transcript levels in GBM, compared with that observed in normal tissues and low-grade gliomas. This study also shows that overall survival was inversely correlated with HOTAIR levels in GBM [44]. In another study, Zhou et al. [45] measured HOTAIR levels in gliomas and normal brain tissues and found a higher expression of HOTAIR in GBM tissues than in low-grade gliomas and normal brain tissues. Thus HOTAIR has potential for use as a serum biomarker for GBM diagnosis and prognosis since it appears to enable a correlation between HOTAIR expression and glioma grade.

Fig. 3.

HOTAIR role in epigenetic modifications. PRC2 is a target that HOTAIR can bind to and induce changes. The PRC2 complex then induces lysine methylation on histone H3. This H3K27-methylation is a form of gene silencing and is induced by histone methyltransferase EZH2, a component of PRC2. The LSD1 demethylase creates a repressor complex with REST and CoREST, that together have a pivotal role in gene silencing through mediating enzymatic demethylation of H3K4. HOTAIR interacts with both PRC2 and LSD1. The HOTAIR-PRC2-LSD1 complex attaches to the promoters of many tumor suppressor genes enabling HOTAIR to silence those genes. This histone methylation and demethylation is a dynamic process of epigenetic modification that regulates gene transcription, chromatin packaging, and cellular differentiation. Abbreviations: HOTAIR, HOX transcript antisense RNA; lncRNA, long non-coding RNA; PRC2, polycomb repressive complex 2; SUZ12, suppressor of zeste 12 protein homolog; EED, embryonic ectoderm development; EZH2, enhancer of zeste homolog 2; REST, RE1-silencing transcription factor; LSD1, lysine-specific histone demethylase 1; coREST, REST corepressor 1; H3K27me3, trimtethylated lysine 27 of histone H3; H3K4deme, demethylated lysine 4 of histone H3.

HOTAIR as a serum diagnostic and prognostic glioblastoma biomarker

Circulating serum HOTAIR was found to be significantly greater in GBM patients than in normal controls, using real-time quantitative reverse transcription PCR (qRT-PCR), as reported by Tan et al. [46]. This finding thus indicates the potential for using HOTAIR as a biomarker to diagnose GBM. Further supporting this potential is the finding by Shen et al. [47] that high levels of HOTAIR were associated with a 2.04-fold decrease in survival.

2.2 Long noncoding RNA nuclear-enriched abundant transcript 1 (lncRNA NEAT1) as serum diagnostic biomarkers

Another mechanism by which lncRNAs can indirectly alter gene expression is by acting as miRNA sponges or as competitive endogenous RNAs (ceRNA) by binding to miRNAs [48]. A single miRNA molecule can directly affect the expression of hundreds of genes by targeting hundreds of mRNAs so miRNAs can repress protein synthesis [49, 50]. As shown in Fig. 4, lncRNA binding to miRNAs can de-repress the expression of all target genes that were acted upon tumor suppressor genes or oncogenes [51].

Fig. 4.

Long noncoding RNA acts as a competitive endogenous RNA. LncRNAs (1) bind to miRNAs. This prevents miRNAs (2) from binding to mRNA (3) thus allowing mRNA translation (4).

LncRNA NEAT1 acts as a ceRNA, as discussed above. LncRNA NEAT1 binds to many miRNAs that have a suppressive effect on glioma cells growth and proliferation, including miR-139-5p, miR-132, miR-128-3p, miR-98-5p, and miR-107 [52, 53, 54, 55, 56, 57]. Subsequently, this leads to tumor cell proliferation and metastasis. Using qRT-PCR to measure NEAT1 expression in cultured tumor tissues from patients with gliomas and non-tumorous tissues, He et al. [58] found significantly higher NEAT 1 expression in glioma tissues. They also reported larger tumor sizes, higher WHO grade, and recurrence were strongly correlated with high levels of NEAT1 upregulation. Further, unfavorable prognoses were associated with patients receiving postoperative chemoradiotherapy with high NEAT1 expression [58]. This suggests a role for lncRNA NEAT1 expression in high-grade gliomas cell proliferation and metastasis, and hence the potential for novel treatments by targeting lncRNA NEAT1. Clinically, Wu et al. [59] found high expression of lncRNA NEAT1 in the plasma of GBM patients.

2.3 LncRNA growth arrest-specific transcript 5 (GAS5) as a serum diagnostic and prognostic biomarker

LncRNA GAS5 gene is located on chromosome 1q12.1 [60]. LncRNA GAS5 is considered an antioncogenic lncRNA and is downregulated in many tumors including ovarian and breast cancers, non-small cell lung cancers, large B‑cell lymphoma, and gastric and laryngeal cancers [60, 61, 62, 63, 64, 65]. In contrast, lncRNA GAS5 acts as an oncogenic lncRNA and is overexpressed in other tumors such as hepatocellular carcinoma and cholangiocarcinoma [66, 67]. LncRNA GAS5 suppresses glioma proliferation and growth by acting as a ceRNA through targeting multiple miRNAs including miRNA-222, miRNA-10b, and miR-196a-5p [68, 69, 70].

In GBM tissues, Toraih et al. [71] found that lncRNA GAS5 was significantly under-expressed. Clinically, Shen et al. [47] found that high serum levels of GAS5 were associated with a 56% decrease in the likelihood of death in GBM patients. This indicates the possibility of using LncRNA GAS5 as a prognostic and diagnostic biomarker for GBM patients.

2.4 LncRNA survival associated mitochondrial melanoma-specific oncogenic non-coding RNA (lncRNA SAMMSON) as a serum diagnostic biomarker

LncRNA SAMMSON gene is located on chromosome 3p13–3p14 and is known to act as an oncogene in different tumors, such as melanoma, liver tumor, gastric cancer, papillary thyroid carcinoma, and oral squamous cell carcinoma [72, 73, 74, 75, 76]. In GBM tissues and cells, Ni et al. [77] detected high expression of lncRNA SAMMSOM. SAMMSON acts as a ceRNA through binding to miRNA-622. Through this mechanism, it increases GBM cell proliferation [78]. Clinically, Xie et al. [78] found that lncRNA SAMMSON was overexpressed in GBM patient plasma comparing to normal controls.

3. GBM-associated lncRNAs as potential diagnosis and prognosis serum biomarkers

We recognize that many lncRNAs have been examined in GBM biopsy specimens and cell cultures that show a significant correlation between tumor progression and patient prognosis. However, clinical evidence for these lncRNAs as serum biomarkers is lacking. For instance, HOXA11-AS is an example of a potentially promising lncRNA biomarker. HOXA11-AS is another HOX family member related to GBM. Like other HOX genes that cause tumor formation when inappropriately expressed or dysregulated. HOXA11-AS expression plays a role in tumorigeneses including non-small cell lung cancers, osteosarcoma, and hepatocellular carcinoma [79, 80, 81]. Li et al. [82] found that miR-140-5p has a role in inhibition proliferation and induction apoptosis of Wilms’ tumor. Others have also reported that glioma cell proliferation and invasion are inhibited effectively by overexpressed miR-140-5p levels [83]. This indicates a protective role for miR-140-5p against tumorigenesis. Cui et al. [84] suggested that HOXA11-AS attaches to miR-140-5p and prevents its protective role. Subsequently, this leads to glioma cells proliferation and invasion. Wang et al. [85] found that HOXA11-AS is overexpressed in GBM tissues and high levels of HOXA11-AS are inversely associated with overall survival of GBM patients, thus suggesting that HOXA11-AS could be a prognostic biomarker. Bountali et al. [86] showed that the long-term survival of GBM cells is decreased when lncRNA MIAT is downregulated. This shows a role for lncRNA MIAT in GBM growth and progression. Through binding to miRNA-182, LncRNA UCA1 promotes GBM cells proliferation and invasion by de-repressing expression of PFKFB2 which is responsible of increasing glycolysis in GBM cells [87]. Another lncRNA that plays an important role in GBM growth through increasing glycolysis in GBM cells is lncRNA SNHG9. LncRNA SNHG9 increases Wnt2 expression by sponging miRNA-199a-5p. De-repressed Wnt2 increases aerobic glycolysis in GBM cells which thus increases GBM cell growth and proliferation capacity [88]. By producing miR-22-3p and miR-22-5p, lncRNA MIR22HG promotes GBM cell progression and self-renewal. Han et al. [89] found that MIR22HG silencing leads to loses in miR-22-3p and miR-22-5p, which subsequently leads to the inactivation of Wnt/β-catenin signaling, thus reducing cell proliferation and tumor invasion. Wang et al. [90] found that LncRNA SNHG4 promotes GBM cells proliferation through acting as a ceRNA by binding to miRNA-138. Chai et al. [91] found that lncRNA LINC01579 upregulates EIF4G2 expression in GBM cells by sponging miR-139-5p which subsequently promotes cell proliferation. Table 1 (Ref. [33, 44, 45, 46, 52, 53, 54, 55, 56, 57, 58, 68, 69, 70, 78, 83, 84, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100]) summarizes all the GBM-associated lncRNAs discussed in this review.

Table 1.Aberrant expression of long noncoding RNAs (lncRNA) involved in glioblastoma (GBM).
LncRNA Expression in GBM Role Targets Measured in GBM patient serum References
HOTAIR Upregulated Oncogenic Interacts with the PRC2 complex Yes [44, 45, 46, 92]
NEAT1 Upregulated Oncogenic MiR-139-5p, miR-132, miR-128-3p, miR-98-5p, and miR-107 Yes [52, 53, 54, 55, 56, 57, 58]
GAS5 Downregulated Anti-oncogenic MiRNA-222, miRNA-10b, and miR-196a-5p Yes [68, 69, 70]
SAMMSON Upregulated Oncogenic MiRNA-622 Yes [78]
HOXA11-AS Upregulated Oncogenic MicroRNA-140-5p No [83, 84]
MIAT Upregulated Oncogenic Acting as ceRNA No [86]
UCA1 Upregulated Oncogenic MiRNA-182 No [87]
SNHG9 Upregulated Oncogenic MiRNA-199a-5p No [88]
MIR22HG Upregulated Oncogenic MiR-22-3p and miR-22-5p No [89]
SNHG4 Upregulated Oncogenic MiR-138 No [90]
LINC01579 Upregulated Oncogenic MiR-139-5p No [91]
SBF2-AS1 Upregulated Oncogenic MiR-151a-3p Yes [33]
MALAT 1 Upregulated Oncogenic MiRNA‐101 and miRNA 203 Yes [93, 94]
TP73-AS1 Upregulated Oncogenic Regulation of the expression of metabolism- related genes and ALDH1A1 No [95]
OIP5-AS1 Upregulated Oncogenic MiRNA-129-5p No [96]
CRNDE Upregulated Oncogenic PI3K/Akt/mTOR pathway and ABCG2 expression No [97]
LINC00511 Upregulated Oncogenic MiRNA-126-5p No [98]
TUSC7 Downregulated Anti-oncogenic MiRNA-10a No [99]
AC003092.1 Downregulated Anti-oncogenic MiRNA-195 No [100]
4. LncRNAs as biomarkers for responsiveness of glioblastoma to anti-cancer drugs

A major barrier to chemotherapy efficacy with GBM is that tumors become metastatic and/or chemoresistant in a large portion of patients following chemotherapy [101, 102]. The absence of effective therapies for GBM makes it crucial to understand the pathophysiology of chemoresistance. Of interest, there are specific lncRNA and miRNA associations with GBM chemoresistance. The lncRNA mechanism for chemoresistance development is largely unknown, hence its study could provide new therapeutic targets that can reverse GBM chemoresistance. Additional clinical research could also confirm the association between high levels of identified lncRNAs and GBM chemoresistance so these lncRNAs can also be examined as potential biomarkers for GBM chemoresistance, thus protecting those patients from ineffective and thus unnecessary and harmful chemotherapies.

4.1 HOTAIR as a biomarker for GBM responsiveness to Bromodomain and Extraterminal (BET) Inhibitors

By binding to the HOTAIR promoter, Bromodomain Containing 4 (BRD4) protein controls HOTAIR levels and increases its expression. Pastori et al. [92] found that by using small molecule BET inhibitors, which are promising epigenetic modulators currently used in clinical trials, BRD4 activity can be inhibited to decrease BRD4 binding at the HOTAIR promoter. HOTAIR levels have been shown to decrease both in vitro and in vivo [92]. This inhibition of HOTAIR transcription is crucial to induce cell cycle arrest in GBM cells. Pastori et al. [92] suggested that HOTAIR can be used as a biomarker for the responsiveness of GBM cells to BET inhibitors. More research to examine the correlation between serum HOTAIR and GBM resistance to BET inhibitors is thus warranted.

4.2 Long noncoding RNAs as serum biomarkers for GBM responsiveness to temozolomide (TMZ)

TMZ is an alkylating agent that works by attaching an alkyl group (CnH2n+1) to the purine bases of DNA [103]. TMZ is metabolized finally to the electrophilic alkylating methyldiazonium cation that acts as a donor of a methyl group on the O6 position of guanine. Although the majority of DNA-methyl adducts are on the N7 of guanine and the N3 of adenine, the O6 position methylations accounts for about 5% of DNA adducts and are primarily responsible for the cytotoxic effects of TMZ [104]. The subsequent methyl group transferring to the O6 position of guanine forms O6-methylguanine lesion leading to DNA double-strand breaks. As a result, apoptosis and/or autophagy occur that cause cell death [105, 106]. TMZ was approved by the U.S. Food and Drug Administration for the treatment of glioblastoma [107]. There are many mechanisms of TMZ resistance for GBM [108], but here we will focus on lncRNAs role in developing TMZ resistance, their potential use as a biomarker for TMZ resistance, and their use as a novel therapeutic agent.

In contrast to bacteria that develop antibiotic resistance by acquiring protein-coding DNA pieces from other bacteria, non-coding genomic pieces make GBM resistant to TMZ. Of interest, by transferring those lncRNAs from chemoresistance GBM tissue or culture to a chemo-sensitive one, the chemo-sensitive tissue becomes chemoresistant [33].

4.2.1 Long noncoding RNA SBF2-AS1 as a serum biomarker for TMZ-resistant glioblastoma

The lncRNA SBF2-AS1 gene is located on chromosome 11p15.1. The proliferation or invasion of many tumor types are associated with altered lncRNA SBF2-AS1 serum levels; Diffuse large B-cell lymphoma, colorectal cancer, hepatocellular carcinoma, serous ovarian carcinoma, clear cell renal cell carcinoma, esophageal squamous cell carcinoma, and lung cancer are associated with increased SBF2-AS1 serum levels [109, 110, 111, 112, 113, 114, 115], while laryngeal squamous cell carcinoma [116] is associated with decreased levels. LncRNA SBF2-AS1 can indirectly alter gene expression by acting as a ceRNA. LncRNA SBF2-AS1 binds to protective miRNAs, thus inducing tumor proliferation and invasion [109, 110, 111]. In contrast, lncRNA SBF2-AS1 acts as an anti-oncogene in laryngeal squamous cell carcinoma by targeting a different miRNA associated with laryngeal cancer invasion and migration [116].

In regard to GBM, lncRNA SBF2-AS1 binds to miR-151a-3p which promotes TMZ sensitivity in GBM cells by targeting the XRCC4 gene [33]. Zhang et al. [33] found that overexpression of long non-coding RNA SBF2-AS1 occurs in TMZ-resistant GBM tissues. This overexpression changes the tumor microenvironment and promotes TMZ resistance. Of interest, in lncRNA SBF2-AS1-depleted Rec GBM cells, the cells became more sensitive to TMZ. Thus, exosomal lncSBF2-AS1 in human serum may serve as a possible diagnostic biomarker for TMZ-resistant GBM [33]. This assumption has been confirmed clinically; Zhang et al. [33] found a poor response to TMZ in GBM patients having high levels of lncSBF2-AS1 in serum exosomes.

4.2.2 Long noncoding RNA MALAT 1 as a serum biomarker for TMZ-resistant glioblastoma

LncRNA MALAT1, also called nuclear-enriched abundant transcript 2, is overexpressed in tumor tissues and correlates with prognosis for many tumors, including non-small cell lung cancer, hepatocellular carcinoma, bladder carcinoma, breast cancer, and prostate cancer [117, 118, 119, 120, 121, 122]. With respect to gliomas, lncRNA MALAT1 promotes growth and progression by acting as a ceRNA sponge for miRNA-613 and miRNA-124 [123, 124].

With respect to chemoresistance, lncRNA MALAT1 has been found to have a major role in developing chemoresistance for osteosarcomas, chronic myeloid leukemia, ovarian cancer, and hepatocellular carcinoma [125, 126, 127, 128]. MALAT1 promoted the TMZ resistance through suppressing miRNA-101 and miRNA 203 signaling pathways in GBM cells [93, 94]. Clinically, Chen et al. [94] found that high serum MALAT1 levels are correlated with TMZ chemoresistance. This indicates the possibility for lncRNA MALAT1 as a biomarker for GBM chemoresistance.

4.3 Other GBM-associated lncRNAs that need to be measured in GBM patients’ serum to act as noninvasive serum biomarkers for TMZ chemoresistance

Mazor et al. [95] found that lncRNA TP73-AS1 promotes TMZ resistance in GBM cancer stem cells and that there is a significant overexpression of lncRNA TP73-AS1 in GBM biopsies. This suggests a potential for this lncRNA to be used as a biomarker to predict TMZ chemoresistance. LncRNA OIP5-AS1 acts as a ceRNA by binding to miRNA-129-5p. Since miRNA-129-5p targets and downregulates IGF2BP2, lncRNA OIP5-AS1 can cause TMZ sensitivity if it is inhibited [96]. Thus, targeting this lncRNA could provide a novel therapeutic pathway for treating GBM patients with TMZ resistance. Another lncRNA that can cause TMZ sensitivity when it is knocked down is lncRNA CRNDE [97]. In GBM cells, LncRNA LINC00511 can promote TMZ chemoresistance by sponging miRNA-126-5p [98]. In contrast to those lncRNAs, there are other lncRNAs which have a protective role and induce chemosensitivity. For instance, lncRNA TUSC7 targets miRNA-10a in GBM cells which inhibits TMZ resistance [99]. LncRNA AC003092.1 is another lncRNA that promotes TMZ chemosensitivity in GBM cells which it does through binding to miRNA-195 [100].

5. Conclusions

In contrast to other invasive modalities of GBM diagnosis, circulating genetic biomarkers are easier, faster, and less costly. These represent an interesting potential method for diagnosis, treatment, prognosis, and avoiding chemoresistance when chemotherapy is unneeded. LncRNAs have shown a potential strong correlation with GBM diagnosis, prognosis, and grading. Furthermore, this suggests other mechanisms for chemoresistance that require further elucidation. There is thus great hope that these biomarkers can be used clinically in the future to decrease the need for invasive diagnostic procedures, to help patients with chemoresistance in their diagnosis and treatment, and to target those lncRNA that cause chemoresistance. LncRNAs have been proven to have a significant value in diagnostic and prognostic purposes. Robust experimental and clinical research should be utilized to determine the most sensitive and highly specific markers for correlating between high plasma levels with GBM type and virulence and to examine the possibility of using lncRNAs as easy, rapid, and noninvasive methods for GBM diagnosis and treatment.

Author contributions

AME performed the research and wrote the manuscript. SLG and AK provided reviewing, editing. MKB provided help, advice, and editing. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This research received no external funding.

Conflict of interest

The authors declare no conflict of interest.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References
[1]
Ostrom QT, Bauchet L, Davis FG, Deltour I, Fisher JL, Langer CE, et al. The epidemiology of glioma in adults: a “state of the science” review. Neuro-Oncology. 2014; 16: 896–913.
[2]
Goodenberger ML, Jenkins RB. Genetics of adult glioma. Cancer Genetics. 2012; 205: 613–621.
[3]
Ohgaki H, Kleihues P. Genetic pathways to primary and secondary glioblastoma. the American Journal of Pathology. 2007; 170: 1445–1453.
[4]
Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathologica. 2016; 131: 803–820.
[5]
Campos B, Olsen LR, Urup T, Poulsen HS. A comprehensive profile of recurrent glioblastoma. Oncogene. 2017; 35: 5819–5825.
[6]
Razavi S, Lee KE, Jin BE, Aujla PS, Gholamin S, Li G. Immune Evasion Strategies of Glioblastoma. Frontiers in Surgery. 2016; 3: 11.
[7]
Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJB, Janzer RC, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. The Lancet Oncology. 2009; 10: 459–466.
[8]
Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. The New England Journal of Medicine. 2005; 352: 987–996.
[9]
Wu L, Qu X. Cancer biomarker detection: recent achievements and challenges. Chemical Society Reviews. 2016; 44: 2963–2997.
[10]
Carninci P, Kasukawa T, Katayama S, Gough J, Frith MC, Maeda N, et al. The transcriptional landscape of the mammalian genome. Science. 2005; 309: 1559–1563.
[11]
Johnson JM, Edwards S, Shoemaker D, Schadt EE. Dark matter in the genome: evidence of widespread transcription detected by microarray tiling experiments. Trends in Genetics. 2005; 21: 93–102.
[12]
Kapranov P, Cheng J, Dike S, Nix DA, Duttagupta R, Willingham AT, et al. RNA Maps Reveal New RNA Classes and a Possible Function for Pervasive Transcription. Science. 2007; 316: 1484–1488.
[13]
Wahlestedt C. Targeting long non-coding RNA to therapeutically upregulate gene expression. Nature Reviews. Drug Discovery. 2013; 12: 433–446.
[14]
Venkatesh T, Suresh PS, Tsutsumi R. Non-coding RNAs: Functions and applications in endocrine-related cancer. Molecular and Cellular Endocrinology. 2016; 416: 88–96.
[15]
Ma L, Bajic VB, Zhang Z. On the classification of long non-coding RNAs. RNA Biology. 2014; 10: 925–933.
[16]
Hangauer MJ, Vaughn IW, McManus MT. Pervasive transcription of the human genome produces thousands of previously unidentified long intergenic noncoding RNAs. PLoS Genet. 2013; 9: e1003569.
[17]
Gomes AQ, Nolasco S, Soares H. Non-coding RNAs: multi-tasking molecules in the cell. International Journal of Molecular Sciences. 2013; 14: 16010–16039.
[18]
Yadav B, Pal S, Rubstov Y, Goel A, Garg M, Pavlyukov M, et al. LncRNAs associated with glioblastoma: from transcriptional noise to novel regulators with a promising role in therapeutics. Molecular Therapy - Nucleic Acids. 2021; 24: 728–742.
[19]
Hung T, Chang HY. Long noncoding RNA in genome regulation: prospects and mechanisms. RNA Biology. 2010; 7: 582–585.
[20]
Huarte M. The emerging role of lncRNAs in cancer. Nature Medicine. 2015; 21: 1253–1261.
[21]
Kondo Y, Shinjo K, Katsushima K. Long non-coding RNAs as an epigenetic regulator in human cancers. Cancer Science. 2017; 108: 1927–1933.
[22]
Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nature Reviews Genetics. 2009; 10: 155–159.
[23]
Sun W, Yang Y, Xu C, Guo J. Regulatory mechanisms of long noncoding RNAs on gene expression in cancers. Cancer Genetics. 2017; 216-217: 105–110.
[24]
Kumarswamy R, Bauters C, Volkmann I, Maury F, Fetisch J, Holzmann A, et al. Circulating long noncoding RNA, LIPCAR, predicts survival in patients with heart failure. Circulation Research. 2014; 114: 1569–1575.
[25]
Lorenzen JM, Schauerte C, Kielstein JT, Hübner A, Martino F, Fiedler J, et al. Circulating long noncoding RNATapSaki is a predictor of mortality in critically ill patients with acute kidney injury. Clinical Chemistry. 2015; 61: 191–201.
[26]
Yang Y, Cai Y, Wu G, Chen X, Liu Y, Wang X, et al. Plasma long non-coding RNA, CoroMarker, a novel biomarker for diagnosis of coronary artery disease. Clinical Science. 2015; 129: 675–685.
[27]
Tong Y, Wang X, Zhou X, Liu Z, Yang T, Shi W, et al. Identification of the long non-coding RNA POU3F3 in plasma as a novel biomarker for diagnosis of esophageal squamous cell carcinoma. Molecular Cancer. 2015; 14: 3.
[28]
Zhou X, Yin C, Dang Y, Ye F, Zhang G. Identification of the long non-coding RNA H19 in plasma as a novel biomarker for diagnosis of gastric cancer. Scientific Reports. 2015; 5: 11516.
[29]
Zhang J, Han L, Bao Z, Wang Y, Chen L, Yan W, et al. HOTAIR, a cell cycle–associated long noncoding RNA and a strong predictor of survival, is preferentially expressed in classical and mesenchymal glioma. Neuro-Oncology. 2013; 15: 1595–1603.
[30]
Xu CH, Xiao LM, Liu Y, Chen LK, Zhang SY, Zeng EM, et al. The lncRNA HOXA11-AS promotes glioma cell growth and metastasis by targeting miR-130a-5p/HMGB2. European Review for Medical and Pharmacological Sciences. 2019; 23: 241–252.
[31]
Chen Q, Cai J, Wang Q, Wang Y, Liu M, Yang J, et al. Long Noncoding RNA NEAT1, Regulated by the EGFR Pathway, Contributes to Glioblastoma Progression through the WNT/β-Catenin Pathway by Scaffolding EZH2. Clinical Cancer Research. 2018; 24: 684–695.
[32]
Peng Z, Liu C, Wu M. New insights into long noncoding RNAs and their roles in glioma. Molecular Cancer. 2018; 17: 61.
[33]
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. Journal of Experimental & Clinical Cancer Research. 2019; 38: 166.
[34]
Baspinar Y, Elmaci I, Ozpinar A, Altinoz MA. Long non-coding RNA MALAT1 as a key target in pathogenesis of glioblastoma. Janus faces or Achilles’ heal? Gene. 2020; 739: 144518.
[35]
Yang C, Wang L, Sun J, Zhou J, Tan Y, Wang Y, et al. Identification of long non-coding RNA HERC2P2 as a tumor suppressor in glioma. Carcinogenesis. 2019; 40: 956–964.
[36]
Wang W, He X, Zheng Z, Ma X, Hu X, Wu D, et al. Serum HOTAIR as a novel diagnostic biomarker for esophageal squamous cell carcinoma. Molecular Cancer. 2017; 16: 75.
[37]
Cantile M, Galletta F, Franco R, Aquino G, Scognamiglio G, Marra L, et al. Hyperexpression of HOXC13, located in the 12q13 chromosomal region, in well-differentiated and dedifferentiated human liposarcomas. Oncology Reports. 2013; 30: 2579–2586.
[38]
Petherick A. Genetics: the production line. Nature. 2008; 454: 1042–1045.
[39]
Croce CM. LINCing chromatin remodeling to metastasis. Nature Biotechnology. 2010; 28: 931–932.
[40]
Tsai M, Manor O, Wan Y, Mosammaparast N, Wang JK, Lan F, et al. Long Noncoding RNA as Modular Scaffold of Histone Modification Complexes. Science. 2010; 329: 689–693.
[41]
Zhang Y, Zhang K, Luo Z, Liu L, Wu L, Liu J. Circulating long non-coding HOX transcript antisense intergenic ribonucleic acid in plasma as a potential biomarker for diagnosis of breast cancer. Thoracic Cancer. 2016; 7: 627–632.
[42]
Berrondo C, Flax J, Kucherov V, Siebert A, Osinski T, Rosenberg A, et al. Expression of the long non-coding RNA HOTAIR correlates with disease progression in bladder cancer and is contained in bladder cancer patient urinary exosomes. PLoS ONE. 2016; 11: e0147236.
[43]
Cantile M, Scognamiglio G, Marra L, Aquino G, Botti C, Falcone MR, et al. HOTAIR role in melanoma progression and its identification in the blood of patients with advanced disease. Journal of Cellular Physiology. 2017; 232: 3422–3432.
[44]
Zhang J, Han L, Bao Z, Wang Y, Chen L, Yan W, et al. HOTAIR, a cell cycle-associated long noncoding RNA and a strong predictor of survival, is preferentially expressed in classical and mesenchymal glioma. Neuro-Oncology. 2013; 15: 1595–1603.
[45]
Zhou X, Ren Y, Zhang J, Zhang C, Zhang K, Han L, et al. HOTAIR is a therapeutic target in glioblastoma. Oncotarget. 2015; 6: 8353–8365.
[46]
Tan SK, Pastori C, Penas C, Komotar RJ, Ivan ME, Wahlestedt C, et al. Serum long noncoding RNA HOTAIR as a novel diagnostic and prognostic biomarker in glioblastoma multiforme. Molecular Cancer. 2018; 17: 74.
[47]
Shen J, Hodges TR, Song R, Gong Y, Calin GA, Heimberger AB, et al. Serum HOTAIR and GAS5 levels as predictors of survival in patients with glioblastoma. Molecular Carcinogenesis. 2018; 57: 137–141.
[48]
Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi P. A ceRNA Hypothesis: the Rosetta Stone of a Hidden RNA Language? Cell. 2011; 146: 353–358.
[49]
Lu TX, Rothenberg ME. MicroRNA. Journal of Allergy and Clinical Immunology. 2018; 141: 1202–1207.
[50]
Filipowicz W, Bhattacharyya SN, Sonenberg N. Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nature Reviews Genetics. 2008; 9: 102–114.
[51]
Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ, Pandolfi PP. A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature. 2010; 465: 1033–1038.
[52]
Wang H, Yan X, Ji L, Ji X, Wang P, Guo S, et al. MiR-139 Functions as an Antioncomir to Repress Glioma Progression through Targeting IGF-1 R, AMY-1, and PGC-1β. Technology in Cancer Research & Treatment. 2017; 16: 497–511.
[53]
Wu DM, Wang S, Wen X, Han XR, Wang YJ, Fan SH, et al. Long noncoding RNA nuclear enriched abundant transcript 1 impacts cell proliferation, invasion, and migration of glioma through regulating miR-139-5p/ CDK6. Journal of Cellular Physiology. 2019; 234: 5972–5987.
[54]
Zhou K, Zhang C, Yao H, Zhang X, Zhou Y, Che Y, et al. Knockdown of long non-coding RNA NEAT1 inhibits glioma cell migration and invasion via modulation of SOX2 targeted by miR-132. Molecular Cancer. 2018; 17: 105.
[55]
Chen J, Wang H, Wang J, Niu W, Deng C, Zhou M. LncRNA NEAT1 Enhances Glioma Progression via Regulating the miR-128-3p/ITGA5 Axis. Molecular Neurobiology. 2021; 58: 5163–5177.
[56]
Li Y, Wang X, Zhao Z, Shang J, Li G, Zhang R. LncRNA NEAT1 promotes glioma cancer progression via regulation of miR-98-5p/BZW1. Bioscience Reports. 2021; 41: BSR20200767.
[57]
Zhen Y, Nan Y, Guo S, Zhang L, Li G, Yue S, et al. Knockdown of NEAT1 repressed the malignant progression of glioma through sponging miR‐107 and inhibiting CDK14. Journal of Cellular Physiology. 2019; 234: 10671–10679.
[58]
He C, Jiang B, Ma J, Li Q. Aberrant NEAT1 expression is associated with clinical outcome in high grade glioma patients. Acta Pathologica, Microbiologica, Et Immunologica Scandinavica. 2016; 124: 169–174.
[59]
Wu X, Jiang T, Huang R, Xiao X. LncRNA GAS8-AS1 downregulates lncRNA NEAT1 to inhibit glioblastoma cell proliferation. Brain and Behavior. 2021; 11: e02128.
[60]
Zhao H, Yu H, Zheng J, Ning N, Tang F, Yang Y, et al. Lowly-expressed lncRNA GAS5 facilitates progression of ovarian cancer through targeting miR-196-5p and thereby regulating HOXA5. Gynecologic Oncology. 2018; 151: 345–355.
[61]
Kanabe BO, Ozaslan M, Aziz SA, Al-Attar MS, Kılıç İH, Khailany RA. Expression patterns of LncRNA-GAS5 and its target APOBEC3C gene through miR-103 in breast cancer patients. Cellular and Molecular Biology. 2021; 67: 5–10.
[62]
Fu Y, Liu L, Zhan J, Zhan H, Qiu C. LncRNA GAS5 expression in non-small cell lung cancer tissues and its correlation with Ki67 and EGFR. American Journal of Translational Research. 2021; 13: 4900–4907.
[63]
Miao Y, Chen X, Qin M, Zhou W, Wang Y, Ji Y. lncRNA GAS5, as a ceRNA, inhibits the proliferation of diffuse large B‑cell lymphoma cells by regulating the miR‑18a‑5p/RUNX1 axis. International Journal of Oncology. 2021; 59: 94.
[64]
Wei MF, Gu ZS, Zheng LL, Zhao MX, Wang XJ. Long non-coding RNA GAS5 promotes natural killer cell cytotoxicity against gastric cancer by regulating miR-18a. Neoplasma. 2020; 67: 1085–1093.
[65]
Liu W, Zhan J, Zhong R, Li R, Sheng X, Xu M, et al. Upregulation of Long Noncoding RNA_GAS5 Suppresses Cell Proliferation and Metastasis in Laryngeal Cancer via Regulating PI3K/AKT/mTOR Signaling Pathway. Technology in Cancer Research & Treatment. 2021; 20: 153303382199007.
[66]
Tao R, Hu S, Wang S, Zhou X, Zhang Q, Wang C, et al. Association between indel polymorphism in the promoter region of lncRNA GAS5 and the risk of hepatocellular carcinoma. Carcinogenesis. 2015; 36: 1136–1143.
[67]
Li Q, Fu L, Han L, Li S, Zhang Y, Wang J. Long noncoding RNA GAS5 accelerates cholangiocarcinoma progression by regulating hsa-miR-1297. Cancer Management and Research. 2021; 13: 2745–2753.
[68]
Zhao X, Wang P, Liu J, Zheng J, Liu Y, Chen J, et al. Gas5 Exerts Tumor-suppressive Functions in Human Glioma Cells by Targeting miR-222. Molecular Therapy. 2015; 23: 1899–1911.
[69]
Ding Y, Wang J, Zhang H, Li H. Long noncoding RNA-GAS5 attenuates progression of glioma by eliminating microRNA‐10b and Sirtuin 1 in U251 and a172 cells. BioFactors. 2020; 46: 487–496.
[70]
Zhao X, Liu Y, Zheng J, Liu X, Chen J, Liu L, et al. GAS5 suppresses malignancy of human glioma stem cells via a miR-196a-5p/FOXO1 feedback loop. Biochimica Et Biophysica Acta. Molecular Cell Research. 2017; 1864: 1605–1617.
[71]
Toraih EA, Alghamdi SA, El-Wazir A, Hosny MM, Hussein MH, Khashana MS, et al. Dual biomarkers long non-coding RNA GAS5 and microRNA-34a co-expression signature in common solid tumors. PLoS ONE. 2018; 13: e0198231.
[72]
Leucci E, Vendramin R, Spinazzi M, Laurette P, Fiers M, Wouters J, et al. Melanoma addiction to the long non-coding RNA SAMMSON. Nature. 2016; 531: 518–522.
[73]
Li X, Li M, Chen J, Dai H, Wang L, Xiong Y, et al. SAMMSON drives the self-renewal of liver tumor initiating cells through EZH2-dependent Wnt/β-catenin activation. Oncotarget. 2017; 8: 103785–103796.
[74]
Sun SB, Lin SX, Cao HL, Xiao ZQ. Values of long noncoding RNA SAMMSON in the clinicopathologic features and the prognostic implications of human gastric cancer. European Review for Medical and Pharmacological Sciences. 2020; 24: 6080–6087.
[75]
Shao L, Sun W, Wang Z, Dong W, Qin Y. Long noncoding RNA SAMMSON promotes papillary thyroid carcinoma progression through p300/Sp1 axis and serves as a novel diagnostic and prognostic biomarker. IUBMB Life. 2020; 72: 237–246.
[76]
Zheng X, Tian X, Zhang Q, Shi P, Li S. Long non-coding RNA SAMMSON as a novel potential diagnostic and prognostic biomarker for oral squamous cell carcinoma. Journal of Dental Sciences. 2020; 15: 329–335.
[77]
Ni H, Wang K, Xie P, Zuo J, Liu W, Liu C. LncRNA SAMMSON Knockdown Inhibits the Malignancy of Glioblastoma Cells by Inactivation of the PI3K/Akt Pathway. Cellular and Molecular Neurobiology. 2021; 41: 79–90.
[78]
Xie J, Wang X, Liu S, Chen C, Jiang F, Mao K, et al. LncRNA SAMMSON overexpression distinguished glioblastoma patients from patients with diffuse neurosarcoidosis. NeuroReport. 2019; 30: 817–821.
[79]
Chen J, Zhou L, Xu S, Zheng Y, Wan Y, Hu C. Overexpression of lncRNA HOXA11-AS promotes cell epithelial-mesenchymal transition by repressing miR-200b in non-small cell lung cancer. Cancer Cell International. 2017; 17: 64.
[80]
Cui M, Wang J, Li Q, Zhang J, Jia J, Zhan X. Long non-coding RNA HOXA11-as functions as a competing endogenous RNA to regulate ROCK1 expression by sponging miR-124-3p in osteosarcoma. Biomedicine & Pharmacotherapy. 2017; 92: 437–444.
[81]
Yu J, Hong J, Kang J, Liao L, Li C. Promotion of LncRNA HOXA11-as on the proliferation of hepatocellular carcinoma by regulating the expression of LATS1. European Review for Medical and Pharmacological Sciences. 2017; 21: 3402–3411.
[82]
Li JL, Luo P. MiR-140-5p and miR-92a-3p suppress the cell proliferation, migration and invasion and promoted apoptosis in Wilms’ tumor by targeting FRS2. European Review for Medical and Pharmacological Sciences. 2020; 24: 97–108.
[83]
ZHANG R, ZHU JC, HU H, LIN QY, SHAO W, JI TH. MicroRNA-140-5p suppresses invasion and proliferation of glioma cells by targeting glutamate-ammonia ligase (GLUL). Neoplasma. 2020; 67: 371–378.
[84]
Cui Y, Yi L, Zhao J, Jiang Y. Long Noncoding RNA HOXA11-as Functions as miRNA Sponge to Promote the Glioma Tumorigenesis through Targeting miR-140-5p. DNA and Cell Biology. 2017; 36: 822–828.
[85]
Wang Q, Zhang J, Liu Y, Zhang W, Zhou J, Duan R, et al. A novel cell cycle-associated lncRNA, HOXA11-AS, is transcribed from the 5-prime end of the HOXA transcript and is a biomarker of progression in glioma. Cancer Letters. 2016; 373: 251–259.
[86]
Bountali A, Tonge DP, Mourtada-Maarabouni M. RNA sequencing reveals a key role for the long non-coding RNA MIAT in regulating neuroblastoma and glioblastoma cell fate. International Journal of Biological Macromolecules. 2019; 130: 878–891.
[87]
He Z, You C, Zhao D. Long non-coding RNA UCA1/miR-182/PFKFB2 axis modulates glioblastoma-associated stromal cells-mediated glycolysis and invasion of glioma cells. Biochemical and Biophysical Research Communications. 2018; 500: 569–576.
[88]
Zhang H, Qin D, Jiang Z, Zhang J. SNHG9/miR-199a-5p/Wnt2 Axis Regulates Cell Growth and Aerobic Glycolysis in Glioblastoma. Journal of Neuropathology & Experimental Neurology. 2019; 78: 939–948.
[89]
Han M, Wang S, Fritah S, Wang X, Zhou W, Yang N, et al. Interfering with long non-coding RNA MIR22HG processing inhibits glioblastoma progression through suppression of Wnt/β-catenin signalling. Brain. 2020; 143: 512–530.
[90]
Wang X, Tian W, Wu L, Wei Z, Li W, Xu Y, et al. LncRNA SNHG4 regulates miR-138/c-Met axis to promote the proliferation of glioblastoma cells. NeuroReport. 2020; 31: 657–662.
[91]
Chai Y, Xie M. LINC01579 promotes cell proliferation by acting as a ceRNA of miR‐139‐5p to upregulate EIF4G2 expression in glioblastoma. Journal of Cellular Physiology. 2019; 234: 23658–23666.
[92]
Pastori C, Kapranov P, Penas C, Peschansky V, Volmar C, Sarkaria JN, et al. The Bromodomain protein BRD4 controls HOTAIR, a long noncoding RNA essential for glioblastoma proliferation. Proceedings of the National Academy of Sciences. 2015; 112: 8326–8331.
[93]
Cai T, Liu Y, Xiao J. Long noncoding RNA MALAT1 knockdown reverses chemoresistance to temozolomide via promoting microRNA-101 in glioblastoma. Cancer Medicine. 2018; 7: 1404–1415.
[94]
Chen W, Xu X, Li J, Kong K, Li H, Chen C, et al. MALAT1 is a prognostic factor in glioblastoma multiforme and induces chemoresistance to temozolomide through suppressing miR-203 and promoting thymidylate synthase expression. Oncotarget. 2017; 8: 22783–22799.
[95]
Mazor G, Levin L, Picard D, Ahmadov U, Carén H, Borkhardt A, et al. The lncRNA TP73-AS1 is linked to aggressiveness in glioblastoma and promotes temozolomide resistance in glioblastoma cancer stem cells. Cell Death & Disease. 2019; 10: 246.
[96]
Wang X, Li X, Zhou Y, Huang X, Jiang X. Long non-coding RNA OIP5-AS1 inhibition upregulates microRNA-129-5p to repress resistance to temozolomide in glioblastoma cells via downregulating IGF2BP2. Cell Biology and Toxicology. 2021. (in press)
[97]
Zhao Z, Liu M, Long W, Yuan J, Li H, Zhang C, et al. Knockdown lncRNA CRNDE enhances temozolomide chemosensitivity by regulating autophagy in glioblastoma. Cancer Cell International. 2021; 21: 456.
[98]
Lu Y, Tian M, Liu J, Wang K. LINC00511 facilitates Temozolomide resistance of glioblastoma cells via sponging miR‐126‐5p and activating Wnt/β‐catenin signaling. Journal of Biochemical and Molecular Toxicology. 2021; 35: e22848.
[99]
Shang C, Tang W, Pan C, Hu X, Hong Y. Long non-coding RNA TUSC7 inhibits temozolomide resistance by targeting miR-10a in glioblastoma. Cancer Chemotherapy and Pharmacology. 2018; 81: 671–678.
[100]
Xu N, Liu B, Lian C, Doycheva DM, Fu Z, Liu Y, et al. Long noncoding RNA AC003092.1 promotes temozolomide chemosensitivity through miR-195/TFPI-2 signaling modulation in glioblastoma. Cell Death & Disease. 2018; 9: 1139.
[101]
Gerson SL. Clinical relevance of MGMT in the treatment of cancer. Journal of Clinical Oncology. 2002; 20: 2388–2399.
[102]
Taylor JW, Schiff D. Treatment considerations for MGMT-unmethylated glioblastoma. Current Neurology and Neuroscience Reports. 2015; 15: 507.
[103]
National Institute of Diabetes and Digestive and Kidney Diseases. LiverTox: Clinical and research information on drug-induced liver injury. 2012. Available at: http://www.ncbi.nlm.nih.gov/books/NBK547852/ (Accessed: 25 January 2022).
[104]
Newlands ES, Stevens MF, Wedge SR, Wheelhouse RT, Brock C. Temozolomide: a review of its discovery, chemical properties, pre-clinical development and clinical trials. Cancer Treatment Reviews. 1997; 23: 35–61.
[105]
Roos WP, Batista LFZ, Naumann SC, Wick W, Weller M, Menck CFM, et al. Apoptosis in malignant glioma cells triggered by the temozolomide-induced DNA lesion O6-methylguanine. Oncogene. 2007; 26: 186–197.
[106]
Kanzawa T, Bedwell J, Kondo Y, Kondo S, Germano IM. Inhibition of DNA repair for sensitizing resistant glioma cells to temozolomide. Journal of Neurosurgery. 2003; 99: 1047–1052.
[107]
Cohen MH, Johnson JR, Pazdur R. Food and Drug Administration Drug approval summary: temozolomide plus radiation therapy for the treatment of newly diagnosed glioblastoma multiforme. Clinical Cancer Research. 2005; 11: 6767–6771.
[108]
Nakada M, Furuta T, Hayashi Y, Minamoto T, Hamada J. The strategy for enhancing temozolomide against malignant glioma. Frontiers in Oncology. 2012; 2: 98.
[109]
Fu DW, Liu AC. LncRNA SBF2-AS1 promotes diffuse large B-cell lymphoma growth by regulating FGFR2 via sponging miR-494-3p. Cancer Management and Research. 2021; 13: 571–578.
[110]
Chen G, Gu Y, Han P, Li Z, Zhao J, Gao M. Long noncoding RNA SBF2‐AS1 promotes colorectal cancer proliferation and invasion by inhibiting miR‐619‐5p activity and facilitating HDAC3 expression. Journal of Cellular Physiology. 2019; 234: 18688–18696.
[111]
Wu Y, Yu B, Chen W, Ai X, Zhang W, Dong W, et al. Downregulation of lncRNA SBF2-as1 inhibits hepatocellular carcinoma proliferation and migration by regulating the miR-361-5p/TGF-β1 signaling pathway. Aging. 2021; 13: 19260–19271.
[112]
Luan AA, Hou LL, Zhang FY. Silencing of SBF2-AS1 inhibits cell growth and invasion by sponging microRNA-338-3p in serous ovarian carcinoma. The Kaohsiung Journal of Medical Sciences. 2021. (in press)
[113]
Yang X, Zhang Y, Fan H. Downregulation of SBF2-as1 functions as a tumor suppressor in clear cell renal cell carcinoma by inhibiting miR-338-3p-targeted ETS1. Cancer Gene Therapy. 2021; 28: 813–827.
[114]
Zha W, Li X, Tie X, Xing Y, Li H, Gao F, et al. The molecular mechanisms of the long noncoding RNA SBF2-AS1 in regulating the proliferation of oesophageal squamous cell carcinoma. Scientific Reports. 2021; 11: 805.
[115]
Qi H, Wang L, Zhang X, Sun W, Liu J. LncRNA SBF2-AS1 inhibits apoptosis and promotes proliferation in lung cancer cell via regulating FOXM1. Journal of B.U.ON. 2020; 25: 1761–1770.
[116]
Li Y, Tang B, Lyu K, Yue H, Wei F, Xu Y, et al. Low expression of lncRNA SBF2‐AS1 regulates the miR‐302b‐3p/TGFBR2 axis, promoting metastasis in laryngeal cancer. Molecular Carcinogenesis. 2022; 61: 45–58.
[117]
Tripathi V, Ellis JD, Shen Z, Song DY, Pan Q, Watt AT, et al. The nuclear-retained noncoding RNA MALAT1 regulates alternative splicing by modulating SR splicing factor phosphorylation. Molecular Cell. 2010; 39: 925–938.
[118]
Ji P, Diederichs S, Wang W, Böing S, Metzger R, Schneider PM, et al. MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene. 2003; 22: 8031–8041.
[119]
Xie S, Diao L, Cai N, Zhang L, Xiang S, Jia C, et al. MascRNA and its parent lncRNA MALAT1 promote proliferation and metastasis of hepatocellular carcinoma cells by activating ERK/MAPK signaling pathway. Cell Death Discovery. 2021; 7: 110.
[120]
Liang T, Xu F, Wan P, Zhang L, Huang S, Yang N, Wang Y. Malat-1 expression in bladder carcinoma tissues and its clinical significance. American Journal of Translational Research. 2021; 13: 3555–3560.
[121]
Shih CH, Chuang LL, Tsai MH, Chen LH, Chuang EY, Lu TP, et al. Hypoxia-induced MALAT1 promotes the proliferation and migration of breast cancer cells by sponging MiR-3064-5p. Frontiers in Oncology. 2021; 11: 658151.
[122]
Zhang D, Fang C, Li H, Lu C, Huang J, Pan J, et al. Long ncRNA MALAT1 promotes cell proliferation, migration, and invasion in prostate cancer via sponging miR-145. Translational Andrology and Urology. 2021; 10: 2307–2319.
[123]
Su Y, Liang C, Yang Q. LncRNA MALAT1 promotes glioma cell growth through sponge miR-613. Journal of B.U.ON. 2021; 26: 984–991.
[124]
Cheng H, Zhao H, Xiao X, Huang Q, Zeng W, Tian B, et al. Long Non-coding RNA MALAT1 Upregulates ZEB2 Expression to Promote Malignant Progression of Glioma by Attenuating miR-124. Molecular Neurobiology. 2021; 58: 1006–1016.
[125]
Liu C, Han X, Li B, Huang S, Zhou Z, Wang Z, et al. MALAT-1 is associated with the doxorubicin resistance in U-2OS osteosarcoma Cells. Cancer Management and Research. 2021; 13: 6879–6889.
[126]
Luo J, Gao Y, Lin X, Guan X. Systematic analysis reveals a lncRNA-miRNA-mRNA network associated with dasatinib resistance in chronic myeloid leukemia. Annals of Palliative Medicine. 2021; 10: 1727–1738.
[127]
Mao TL, Fan MH, Dlamini N, Liu CL. LncRNA MALAT1 facilitates ovarian cancer progression through promoting chemoresistance and invasiveness in the tumor microenvironment. International Journal of Molecular Sciences. 2021; 22: 10201.
[128]
Cao Y, Zhang F, Wang H, Bi C, Cui J, Liu F, et al. LncRNA MALAT1 mediates doxorubicin resistance of hepatocellular carcinoma by regulating miR-3129-5p/Nova1 axis. Molecular and Cellular Biochemistry. 2021; 476: 279–292.
Share
Back to top