Get 20M+ Full-Text Papers For Less Than $1.50/day. Start a 14-Day Trial for You or Your Team.

Learn More →

Long non-coding RNA in glioma: novel genetic players in temozolomide resistance

Long non-coding RNA in glioma: novel genetic players in temozolomide resistance ANIMAL CELLS AND SYSTEMS 2023, VOL. 27, NO. 1, 19–28 https://doi.org/10.1080/19768354.2023.2175497 Long non-coding RNA in glioma: novel genetic players in temozolomide resistance a# a# b c a,d Jungwook Roh , Mijung Im , JiHoon Kang , BuHyun Youn and Wanyeon Kim a b Department of Science Education, Korea National University of Education, Cheongju-si, Republic of Korea; Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory, Emory University School of Medicine, Atlanta, GA, USA; Department of Biological Sciences, Pusan National University, Busan, Republic of Korea; Department of Biology Education, Korea National University of Education, Cheongju-si, Republic of Korea ABSTRACT ARTICLE HISTORY Received 25 November 2022 Glioma is the most common primary malignant brain tumor in adults and accounts for Revised 15 January 2023 approximately 80% of brain and central nervous system tumors. In 2021, the World Health Accepted 27 January 2023 Organization (WHO) published a new taxonomy for glioma based on its histological features and molecular alterations. Isocitrate dehydrogenase (IDH) catalyzes the decarboxylation of KEYWORDS isocitrate, a critical metabolic reaction in energy generation in cells. Mutations in the IDH genes Isocitrate dehydrogenase; interrupt cell differentiation and serve as molecular biomarkers that can be used to classify long non-coding RNA; gliomas. For example, the mutant IDH is widely detected in low-grade gliomas, whereas the glioma; temozolomide wild type is in high-grade ones, including glioblastomas. Long non-coding RNAs (lncRNAs) are resistance epigenetically involved in gene expression and contribute to glioma development. To investigate the potential use of lncRNAs as biomarkers, we examined lncRNA dysregulation dependent on the IDH mutation status. We found that several lncRNAs, namely, AL606760.2, H19, MALAT1, PVT1 and SBF2-AS1 may function as glioma risk factors, whereas AC068643.1, AC079228.1, DGCR5, FAM13A-AS1, HAR1A and WDFY3-AS2 may have protective effects. Notably, H19, MALAT1, PVT1, and SBF2-AS1 have been associated with temozolomide resistance in glioma patients. This review study suggests that targeting glioma-associated lncRNAs might aid the treatment of glioma. Introduction reclassifies gliomas into adult-type diffuse gliomas, pedi- Gliomas are primary brain tumors derived from neuro- atric-type diffuse LGGs, pediatric-type diffuse HGGs, cir- glial stem cells or neuroglial progenitor cells (Weller cumscribed astrocytic gliomas, and ependymal tumors et al. 2015). The grading of gliomas based on histological (Horbinski et al. 2022). This system also classifies features has been debated for decades by technical gliomas according to isocitrate dehydrogenase (IDH) limitations and different opinions. However, recent mutation status and uses various molecular markers to advances in molecular genetics have revealed novel classify subgroups by their molecular characteristics changes in several tumors and have changed the diag- (Komori 2022). nostic paradigm for tumors of the central nervous IDH is an essential enzyme involved in major meta- system (CNS). Molecular diagnosis was first introduced bolic processes such as the TCA cycle, glutamine in the World Health Organization (WHO) CNS glioma metabolism, adipogenesis, and redox regulation and classification system published in 2016 (Louis et al. has three isoforms, and IDH1 and IDH2 are used as 2016; Louis et al. 2021). According to this system, glioma biomarkers (Koh et al. 2004; Lee et al. 2004; grades 1 and 2 are low-glioma grades (LGGs), and Yan et al. 2009). The most common mutation detected grades 3 and 4 are high-glioma grades (HGGs) (Chen in human IDH1 replaces arginine at position 132 by et al. 2022). In addition, a new classification in the histidine (R132H), whereas the one in human IDH2 WHO published in 2021 incorporates more molecular does arginine at 172 by lysine (R172 K) (Dang et al. alterations into the diagnosis of many tumors and 2009; Yan et al. 2009). Wildtype IDH enzymes convert CONTACT BuHyun Youn bhyoun72@pusan.ac.kr Department of Biological Sciences, Pusan National University, Busandaehak-ro 63beon-gil 2, Geumjeong-gu, Busan 46241, Republic of Korea; Wanyeon Kim wykim82@knue.ac.kr Department of Biology Education, Korea National University of Education, 250 Taeseongtabyeon-ro, Gangnae-myeon, Heungdeok-gu, Cheongju-si, Chungbuk 28173, Republic of Korea Authors contributed equally. © 2023 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. 20 J. ROH ET AL. isocitrate to α-ketoglutarate (α-KG). However, the Li et al. 2021). In this review, to address the issue of mutant IDHs have changed their substrate specificity, TMZ resistance, we summarize the effects of lncRNA converting α-KG to 2-hydroxyglutarate (2-HG). The con- dysregulation according to IDH mutation status in sequent excessive accumulation of 2-HG leads to glioma. glioma development and their malignant progression, associated metabolic alterations, DNA methylation, IDHs as classification biomarkers in glioma and reactive oxygen species (ROS) production (Dang et al. 2009; Lu et al. 2012; Turcan et al. 2012). The IDH is a representative biomarker of glioma classifi- majority of LGG patients have mutations in IDH, and cation, and WHO primarily classifies gliomas according a significant correlation has been reported between to IDH mutation status. IDH is an enzyme that catalyzes IDH mutation and improved overall survival in LGG the reversible oxidative decarboxylation of isocitrate, (Rickert 1987; Youssef and Miller 2020). WHO CNS which results in the production of carbon dioxide and grade 4 glioblastoma can be classified as primary or α-KG in the TCA cycle during glucose metabolism, secondary. Most primary and secondary glioblastomas which in turn, is coupled with the transformation of are of the IDH-wildtype or mutant types, respectively, NAD(P) to NAD(P)H (Zhao et al. 2009). In mammals, and overall survival is better for the secondary type IDH exists in three isoforms: IDH1 is present in the cyto- (Nobusawa et al. 2009; Wick et al. 2009; Sun et al. plasm and peroxisomes, and IDH2 and IDH3, in the mito- 2013). However, diagnoses of glioma based on the chondrial matrix (Leighton et al. 1969). IDH1 and IDH2 mutation status of the IDH genes alone overlook the use NADP as a cofactor to produce α-KG and NADPH, various origins and characteristics of glioma. Therefore, and the generated NADPH protects cells from oxidative an understanding of the molecular mechanism of damage by participating in key ROS scavenging pro- glioma is required for accurate diagnosis and cesses such as biosynthetic reactions and glutathione treatment. disulfide reduction (Hurley et al. 1991; Xu et al. 2004; Long non-coding RNAs (lncRNAs), which do not Itsumi et al. 2015). IDH3 plays a central role in energy encode proteins and are more than 200 nucleotides in production in the TCA cycle by generating α-KG and length, regulate cellular biological processes in cancer NADH using NAD as a cofactor (Reitman and Yan such as cell proliferation, metastasis, invasion, motility, 2010). Both IDH1 and IDH2 form a homodimer, and drug resistance, and their intracellular locations whereas IDH3 holoenzyme is multimeric and assembled reflect their functions (Seo et al. 2019; Seo et al. 2021; with multiple subunits of α, β, and γ encoded by IDH3a, Chen et al. 2022). Nuclear lncRNAs affect gene IDH3b, and IDH3c, respectively (MacDonald et al. 2013). expression at the transcriptional level, regulating In diffuse gliomas, the IDH1-mutant type is the most histone modification, DNA methylation and chromatin common and observed in more than 90% of cases. The remodeling (Marín-Béjar et al. 2013; Zhang et al. 2014; IDH2-mutant type is observed in approximately 10% of Chu et al. 2015; Do et al. 2019). On the other hand, cyto- cases, but the IDH3-mutant type has never been plasmic lncRNAs are competitive endogenous RNAs observed (Waitkus et al. 2018). (ceRNAs) that interact with mRNAs and miRNAs to epi- In cases of IDH1 mutation, the homodimer formed by genetically regulate gene expression at the post-tran- two IDH1-mutant monomers does not have enzymatic scriptional and translational levels. Recent studies activity, but the heterodimer composed of the IDH-wild- suggest that dysregulations of lncRNA can result in the type and mutant monomer has enzymatic activity initiation, progression, and other malignant phenotypes sufficient to convert α-KG to 2-HG (Dang et al. 2009; of glioma (Karreth et al. 2011; Tay et al. 2014; Shao et al. Yang et al. 2010). IDH is one of the most studied genes 2016; Peng et al. 2018; Zhou et al. 2020). in glioma development, and cancers with IDH1 or IDH2 O -methylguanine-DNA methyltransferase (MGMT) mutations produce 10–100 times more 2-HG than methylation, 1p/19q co-deletion, IDH, and epidermal those with the IDH1 or IDH2 wildtype (Ward et al. growth factor receptor have been widely used as 2010; Leeper et al. 2015). IDH1 mutation converts α-KG glioma biomarkers (McNamara et al. 2013;Ludwig to 2-HG, which accumulates in the cytoplasm, and and Kornblum 2017). Recently, it has been demon- thereby releases carbon from the TCA cycle. This cycle strated that lncRNAs might also function as molecular utilizes metabolic reprogramming to compensate for biomarkers in glioma (Peng et al. 2018). For example, fluctuations in metabolic pathways; for example, gluta- lncRNA dysregulation has been shown to be corre- mate dehydrogenase converts glutamine to α-KG, lated with glioma malignancy and associated with which replenishes the TCA cycle (Hausinger 2004; resistance to radiotherapy and temozolomide (TMZ) Loenarz and Schofield 2008; Xu et al. 2011). In addition (Lu et al. 2020;Chae etal. 2021;Kuang et al. 2021; to the metabolic changes, the hypermethylation of ANIMAL CELLS AND SYSTEMS 21 CpG islands is also affected by IDH mutations. The tumor behavior and prognosis. LncRNA AC068643.1 is glioma-specific methylation pattern provides clues more highly expressed in IDH-mutant glioblastomas regarding the pathogenesis of IDH-mutant gliomas. than in the wildtype. The expression levels of DNA methylation is regulated by methyltransferase AC068643.1 are positively correlated with the protein and demethylase. During demethylation, ten-eleven levels of bone morphogenetic protein (BMP) and myos- translocation methylcytosine dioxygenase (TET) con- tatin (MSTN), and it has been experimentally demon- verts 5-methylcytosine to cytosine in an iron- and α- strated that both BMP and MSTN directly stimulate KG-dependent manner. Furthermore, the activity of AC068643.1 expression (Huang et al. 2020). BMP inhibits TET might be inhibited by 2-HG due to its structural simi- cell proliferation in glioblastoma and astrocytic glioma, larity to α-KG (Xu et al. 2011; Lee et al. 2021). Histone and a BMP-like synthetic is considered a non-cytotoxic methylation status is regulated by histone methyltrans- therapeutic that might prevent glioma growth and ferases such as euchromatic histone lysine methyltrans- recurrence. Altogether, these results suggest that ferase 1 (EHMT1), EHMT2, SET nuclear proto-oncogene AC068643.1 can function as a protective lncRNA in glio- (SET), and enhancer of zeste 2 polycomb repressive blastoma patients (González-Gómez et al. 2014; Sach- complex 2 subunit (EZH2), and demethylases such as deva et al. 2019). lysine demethylase 1A (KDM1A) and KDM4A (Chowdh- Differentially expressed lncRNAs were identified ury et al. 2011; Hayward and Cole 2016; Katoh 2016; using GSE107850, a dataset of glioma patients that Rahman et al. 2021). Like TET, histone demethylases underwent surgical excision and TMZ treatment. A such as KDM4 and KDM5 are inhibited by high levels risky lncRNA AL606760.2 and two protective lncRNAs, of 2-HG (Chowdhury et al. 2011). Many studies report FAM13A antisense RNA 1 (FAM13A-AS1) and that histone methylation markers accumulate in AC079228.1, were suggested as predictors of TMZ various cancers harboring a mutant IDH gene. Also, inhi- efficacy in IDH-mutant type LGGs (Li et al. 2021). Further- bition of histone demethylation by 2-HG leads to more, levels of AL606760.2 and Smad2, which mediates impaired cell differentiation, which might be associated transforming growth factor (TGF)-beta signaling, are with the tumorigenesis of IDH-mutant glioma (Lu et al. positively correlated. Thus, AL606760.2 appears to regu- 2012). late intracellular processes such as cell proliferation, IDH is an essential enzyme for energy metabolism apoptosis, and differentiation (Abdel-Wahab et al. associated with the TCA cycle, and mutated IDH causes 2002). On the other hand, FAM13A-AS1 is involved in 2-HG accumulation, hypermethylation, and oxidative decreasing cell proliferation and differentiation, and its damage. Because of these alterations, it is reasonable levels were found to be significantly correlated with to classify glioblastomas by IDH mutational status. The UBR5 levels (an E3 ubiquitin-protein ligase) (Jiang et al. new molecular markers including IDH will help diagnose 2011; Gudjonsson et al. 2012). The two cancer-related and effectively treat gliomas. genes, pre-mRNA processing factor 40 homolog B (PRPF40B) and zinc finger homeobox 3 (ZFHX3), were suggested to be potential target mRNAs of lncRNA LncRNAs and IDH mutations in glioma AC079228.1. PRPF40B is a splicing gene that directly In brain tumors, IDH mutation status is known to be interacts with splicing factor 1 (SF1), and associated associated with a patient’s prognosis of brain tumors with U2 small nuclear RNA auxiliary factor 1 (U2AF1). and with a well-known molecular biomarker of glioma ZFHX3 acts as a transcription factor for regulation of (Turcan et al. 2012). Reportedly, lncRNAs are epigeneti- myogenic and neuronal differentiation and inhibition cally involved in gene expression regulation and treat- of cell cycle progression (Benjamin et al. 2009; Gudb- ment resistance in brain tumors (Mercer et al. 2009). jartsson et al. 2009). Furthermore, accumulating evidence suggests that In another study, lncRNAs differentially expressed in lncRNAs function as ceRNAs that regulate gene IDH-mutant and IDH-wildtype glioma samples obtained expression and alter cell biological properties such as from several CCGA, TCGA, and GSE16011 datasets were cell viability, proliferation, motility, and invasion (Do analyzed using the LNCipedia database (Chen et al. and Kim 2018; Seo et al. 2020). 2020). Multivariate Cox regression analysis of the prog- nostic performances of differentially expressed lncRNAs identified H19 and plasmacytoma variant translocation Dysfunction of lncRNAs according to IDH- 1 (PVT1) as risky lncRNAs and highly accelerated region mutant status 1A (HAR1A) as a protective lncRNA in glioma. LncRNA Glioblastoma can be classified as IDH-wildtype or H19 promotes angiogenesis in gliomas by increasing mutant types, each of which is associated with a distinct the expressions of hypoxia inducible factor 1 subunit 22 J. ROH ET AL. alpha (HIF-1α) and vascular endothelial growth factor highly expressed in IDH-wildtype than in IDH-mutant (VEGF) through targeting miR-138. In glioma, H19 can glioma and its knockdown in glioblastoma upregulates also target miR-342 and upregulate the Wnt5a/β- miR-338-3p, but downregulates EGF-like domain mul- catenin pathway to promote cell proliferation and tiple 7 (EGFL7), thereby inhibiting angiogenesis migration (Liu et al. 2020; Zhou et al. 2022). Also, (Cancer Genome Atlas Research Network et al. 2015; lncRNA PVT1 targets miR-200a to regulate the cell Yu et al. 2017; Zhang et al. 2021). cycle and promote cell proliferation and invasion An analysis of the CGCA database revealed lncRNA (Zhang et al. 2019). On the other hand, lncRNA HAR1A WDFY3 antisense RNA 2 (WDFY3-AS2) is expressed at acts to suppress the progression of diffuse glioma and low levels in IDH-wildtype gliomas. Kaplan-Meier analy- is expressed at low levels in glioma. In addition, glioma sis showed that WDFY3-AS2 overexpression was associ- patients with high PVT1 expression or low HAR1A ated with longer overall survival than low WDFY3-AS2 expression is reported to be prognostic of poor survival expression, and Cox regression analysis showed its in glioma patients. Moreover, the down-regulation of expression was independently correlated with overall PVT1 and up-regulation of HAR1A improved the survival survival. In addition, gene ontology (GO) and gene set of glioma patients that received chemotherapy and enrichment analysis (GSEA) revealed that WDFY3-AS2 radiotherapy. These findings imply that these lncRNAs is involved in the regulation of synaptic transduction, play critical roles in diffuse glioma progression and glutamate receptors, and TNF signaling pathways (Wu that PVT1 and HAR1A should be explored as promising et al. 2018). biomarkers for diagnosis, prognosis, and targeted LncRNA DiGeorge syndrome critical region gene 5 therapy (Zou et al. 2017). (DGCR5) plays cancer-dependent roles. For example, although DGCR5 levels were unrelated to WHO malig- nancy grade in glioma, they were more downregulated LncRNA dysfunction in IDH-wildtype gliomas in IDH-wildtype glioma than in IDH-mutant glioma. Fur- IDH-wildtype gliomas are associated with poor prog- thermore, DGCR5 increased the levels of Smad7 and noses and high CNS grades. Metastasis-associated lung phosphatase and tensin homolog (PTEN) by sponging adenocarcinoma transcript 1 (MALAT1) is an overex- miR-21 and miR-23a, respectively, in glioma cells. It is pressed lncRNA in glioblastoma and promotes cell also reported that Smad7 inhibits the migratory and growth and tumorigenesis. For example, it has been invasive activities of glioma, and PTEN inhibits cell pro- demonstrated that lncRNA MALAT1 promotes the liferation and enhances apoptosis. Patients with low expression of SRY-box transcription factor 2 (SOX2) by DGCR5 expression levels show poorer overall survival suppressing miR-129 to promote glioma stem cell viabi- (He et al. 2020). lity, proliferative abilities, and tumorigenesis (Xiong et al. 2018). In addition, methyltransferase 3 (METTL3), which TMZ resistance by lncRNAs according to IDH upregulates the expression of MALAT1 through N - mutation status methyladenosine (m A) modification, is also associated with poor prognosis. It was also demonstrated that Glioma classification based on histological features and MALAT1 overexpression promotes the malignant IDH mutation status helps with diagnosis and prognosis. glioma phenotype by inducing epithelial–mesenchymal Although advances in techniques, technologies, surgical transition (EMT) and tumor necrosis factor (TNF) signal- resection, chemotherapy, and radiotherapy have signifi- ing through nuclear factor kappa B (NF-κB) activation cantly improved treatment efficacy outcomes fall far (Chang et al. 2021). short of satisfactory. Furthermore, accumulating evi- Furthermore, lncRNA SBF2 antisense RNA 1 (SBF2- dence indicates lncRNA dysregulations are involved in AS1), which plays an oncogenic role in various tumors, glioma chemo- and radioresistance. This review com- may function as a biomarker in diffuse LGG. Kaplan- piles several differentially expressed lncRNAs in IDH- Meier analysis showed that the prognosis of LGG mutant and IDH-wildtype gliomas, and it has been patients with high SBF2-AS1 expression was poorer shown that lncRNAs SBF2-AS1, PVT1, MALAT1, and H19 than those with low expression. Cox regression analysis significantly contribute to the induction of TMZ showed that SBF2-AS1 is an independent prognostic resistance. factor of poorer overall survival in LGG (Zhang et al. TMZ is a lipophilic anticancer drug, which is widely 2021). Most LGGs are IDH mutant type, and only some used to treat brain tumors. TMZ can be readily delivered are IDH-wildtype. However, the molecular and clinical to brain tissues as it crosses the blood–brain barrier and characteristics of IDH-wildtype LGG are similar to those induces cell death by causing single- or double-strand of glioblastoma. It is reported that SBF2-AS1 is more breaks by incorporating mismatched base pairs. ANIMAL CELLS AND SYSTEMS 23 However, more than 50% of glioma patients do not reportedly, its knockdown effectively promotes glioma respond to TMZ, and lncRNAs appear to be involved in cell sensitivity to TMZ. PVT1 and miR-365 expressions TMZ resistance by regulating chemoresistance-associ- are negatively correlated, and the overexpression of ated gene expression, enhancing cell survival, and inhi- miR-365 suppresses TMZ resistance in glioma cells. biting apoptosis (Lee 2016). Furthermore, numerous PVT1 competitively binds to miR-365 and positively studies have suggested that lncRNA might act as a regulates the expression of E74 like ETS transcription ceRNA of miRNA to modulate the expressions of target factor 4 (ELF4), which contributes to the induction of genes (Salmena et al. 2011). The interaction between glioma stemness and TMZ resistance (Bazzoli et al. lncRNA and miRNA might also influence TMZ resistance 2012; Gong et al. 2021). MALAT1 is frequently overex- in glioma cells. It is reported that sponging of miR-151a- pressed in IDH-wildtype gliomas and enhances resist- 3p by lncRNA SBF2-AS1, which is more highly expressed ance to TMZ administration by regulating the in IDH-wildtype than IDH-mutant gliomas, is responsible expressions of miRNAs. Several studies have shown for TMZ resistance in glioma cells (Zhang et al. 2019). that miR-203 contributes to reduced chemoresistance Furthermore the transcription factor, zinc finger E-box in various cancers (Li et al. 2011; Lin et al. 2016; Zhang binding homeobox 1 (ZEB1), might increase SBF2-AS1 et al. 2016). Furthermore, MALAT1 promotes glioma expression by binding to its promotor region, and the cell proliferation and TMZ resistance by decreasing resulting high level of SBF2-AS1 downregulates miR- miR-203 expression and increasing the expression of 151a-3p and the subsequent upregulation of X-ray thymidylate synthase (TS), a potent cancer chemother- repair cross complementing 4 (XRCC4), a target of miR- apy target (Chu et al. 2003; Chen et al. 2017). In addition, 151a-3p. XRCC4 promotes DNA double-strand break MALAT1 downregulates miR-101, consequently increas- repair and thus enhances cell survival and inhibits apop- ing resistance to TMZ in glioma cells by increasing glyco- tosis in glioma cells in response to TMZ treatment. Fur- gen synthase kinase 3β (GSK-3β) levels. Also, increased thermore, it has been shown that SBF2-AS1 depletion GSK-3β expression might be associated with resistance reduces the level of XRCC4, delays the repair of TMZ- to TMZ and poor prognosis in glioma patients by induced DNA damage, and increases glioma cell sensi- decreasing MGMT promoter methylation and thus upre- tivity to TMZ. PVT1 is a known oncogene in various gulation of MGMT (Pyko et al. 2013; Tian et al. 2016; Cai cancers, including glioma. It is expressed at higher et al. 2018). MALAT1 also regulates TMZ resistance by levels in IDH-mutant than in IDH-wildtype gliomas and modulating multidrug resistance-related genes. The Figure 1. Mechanisms of lncRNAs inducing TMZ resistance. H19 is involved in induction of glioma cell viability and survival through upregulation of MDR1, MRP1, and ABCG2 and activation of NF-κB signaling, responsible for induction of TMZ resistance (Jiang et al. 2016; Duan et al. 2018). MALAT1 is associated with glioma cell proliferation, viability, and invasion through regulation of ceRNA net- works with miRNAs, upregulation of multidrug resistance-related genes, and regulation of ZEB1, which might contribute to glioma TMZ resistance (Chu et al. 2003; de Cremoux et al. 2007; Pyko et al. 2013; Tian et al. 2016; Chen et al. 2017; Li et al. 2017; Cai et al. 2018; Dong et al. 2019). PVT1 and SBF2-AS1 play oncogenic roles in induction of glioma stemness and cell survival, respectively, through regulation of ceRNA networks, resulting in TMZ resistance in glioma cells (Bazzoli et al. 2012; Zhang et al. 2019; Gong et al. 2021). 24 J. ROH ET AL. Figure 2. Therapeutic strategies targeting lncRNAs. Downregulation of risky lncRNAs such as AL606760.2 (Li et al. 2021), H19 (Chen et al. 2020), PVT1 (Chen et al. 2020), MALAT1 (Xiong et al. 2018), and SBF2-AS1 (Zhang et al. 2021) and upregulation of protective lncRNAs such as AC068643.1 (Huang et al. 2020), AC079228.1 (Li et al. 2021), FAM13A-AS1 (Li et al. 2021), HAR1A (Chen et al. 2020), DGCR5 (He et al. 2020), and WDFY3-AS2 (Wu et al. 2018) might help predict prognosis in glioma. downregulation of MALAT1 reduces the mRNA plus drug treatment. However, the discovery of expression levels of multidrug resistance-related pro- lncRNAs, known to induce TMZ resistance, does not teins such as multidrug resistance protein 1 (MDR1), completely explain the difficulties of treating patients multidrug resistance protein 5 (MRP5), and lung–resist- with TMZ-resistant glioma. Nonetheless, targeting ance related protein 1 (LRP1), which are important regu- these lncRNAs is a promising way of reducing TMZ lators of chemoresistance, and also reduces glioma cell resistance. sensitivity to TMZ. In addition, MALAT1 is involved in EMT induction and thus regulates glioblastoma cell sen- Conclusions sitivity to TMZ. The downregulation of MALAT1 decreases TMZ resistance by reducing the expression The dysregulation of lncRNA contributes to the develop- level of ZEB1, an EMT-related protein that increases ment of several types of cancer, and lncRNA promotes cancer cell motility and invasiveness (de Cremoux et al. glioma development and induces chemo- and radiore- 2007; Li et al. 2017; Dong et al. 2019). H19 is highly sistance. IDH mutation status is a widely-accepted bio- expressed in IDH-mutant gliomas and involved in TMZ marker for glioma classification, but additional markers resistance by regulating multidrug resistance genes, are desired to determine effective treatment strategies. and its levels are elevated in TMZ-resistant glioma This review focuses on risky and protective lncRNAs cells. Inhibition of H19 downregulates the expressions according to IDH mutation status and the mechanisms of several multidrug resistance genes such as MDR1, involved in TMZ resistance. We suggest five lncRNAs MRP1, and ATP-binding cassette super-family G acting as risk factors, namely, AL606760.2, H19, member 2 (ABCG2) in TMZ-resistant cells at the mRNA MALAT1, PVT1, and SBF2-AS1, and another six protective and protein levels, which suggests H19 plays an impor- lncRNAs, that are AC068643.1, AC079228.1, DGCR5, tant role in the induction of TMZ resistance in glioma FAM13A-AS1, HAR1A, and WDFY3-AS2. The risky H19, cells (Jiang et al. 2016). Furthermore, oxidative stress MALAT1, PVT1, and SBF2-AS1 increase TMZ resistance induces H19 expression and thus increases cell survival (Figure 2). Identifying these risky or protective lncRNAs and viability by activating NF-κB signaling, which accom- should help predict prognosis in glioma. Determining panies the acquisition of TMZ resistance by glioma cells the dysregulation statuses of lncRNAs associated with (Duan et al. 2018). TMZ resistance should aid the development of promis- In glioma, increased TMZ resistance, conferred by the ing pharmacological targets. Therefore, therapeutic upregulations of risky lncRNAs such as H19, MALAT1, strategies targeting the dysregulation of lncRNA accord- PVT1, and SBF2-AS1 (Figure 1), is likely to play a key ing to IDH mutation status will help improve the prog- role in the therapeutic efficacy of surgical resection noses of glioma patients. ANIMAL CELLS AND SYSTEMS 25 Chowdhury R, Yeoh KK, Tian Y-M, Hillringhaus L, Bagg EA, Rose Disclosure statement NR, Leung IKH, Li XS, Woon ECY, Yang M, et al. 2011. The No potential conflict of interest was reported by the author(s). oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases. EMBO Rep. 12(5):463–469. doi:10.1038/ embor.2011.43. Chu C, Zhang QC, da Rocha ST, Flynn RA, Bharadwaj M, References Calabrese JM, Magnuson T, Heard E, Chang HY. 2015. Systematic discovery of xist RNA binding proteins. Cell. Abdel-Wahab N, Wicks SJ, Mason RM, Chantry A. 2002. Decorin 161(2):404–416. doi:10.1016/j.cell.2015.03.025. suppresses transforming growth factor-beta-induced Chu E, Callender MA, Farrell MP, Schmitz JC. 2003. Thymidylate expression of plasminogen activator inhibitor-1 in human synthase inhibitors as anticancer agents: from bench to mesangial cells through a mechanism that involves Ca2 bedside. Cancer Chemother Pharmacol. 52(Suppl 1):S80– +-dependent phosphorylation of Smad2 at serine-240. S89. doi:10.1007/s00280-003-0625-9. Biochem J. 362(Pt 3):643–649. doi:10.1042/0264- Dang L, White DW, Gross S, Bennett BD, Bittinger MA, 6021:3620643. Driggers EM, Fantin VR, Jang HG, Jin S, Keenan MC, Bazzoli E, Pulvirenti T, Oberstadt MC, Perna F, Wee B, Schultz N, et al. 2009. Cancer-associated IDH1 mutations produce 2- Huse JT, Fomchenko EI, Voza F, Tabar V, et al. 2012. MEF pro- hydroxyglutarate. Nature. 462(7274):739–744. doi:10.1038/ motes stemness in the pathogenesis of gliomas. Cell Stem nature08617. Cell. 11(6):836–844. doi:10.1016/j.stem.2012.09.012. de Cremoux P, Jourdan-Da-Silva N, Couturier J, Tran-Perennou Benjamin EJ, Rice KM, Arking DE, Pfeufer A, van Noord C, Smith C, Schleiermacher G, Fehlbaum P, Doz F, Mosseri V, Delattre AV, Schnabel RB, Bis JC, Boerwinkle E, Sinner MF, et al. 2009. O, Klijanienko J, et al. 2007. Role of chemotherapy resistance Variants in ZFHX3 are associated with atrial fibrillation in genes in outcome of neuroblastoma. Pediatr Blood Cancer. individuals of European ancestry. Nat Genet. 41(8):879– 48(3):311–317. doi:10.1002/pbc.20853. 881. doi:10.1038/ng.416. Do H, Kim D, Kang J, Son B, Seo D, Youn H, Youn B, Kim W. 2019. Cancer Genome Atlas Research Network, Brat DJ, Verhaak TFAP2C increases cell proliferation by downregulating RGW, Aldape KD, Yung WKA, Salama SR, Cooper LAD, GADD45B and PMAIP1 in non-small cell lung cancer cells. Rheinbay E, Miller CR, Vitucci M, et al. 2015. Biol Res. 52(1):35. doi:10.1186/s40659-019-0244-5. Comprehensive, integrative genomic analysis of diffuse Do H, Kim W. 2018. Roles of oncogenic long non-coding RNAs lower-grade gliomas. N Engl J Med. 372(26):2481–2498. in cancer development. Genomics Inform. 16(4):e18. doi:10. doi:10.1056/NEJMoa1402121. 5808/GI.2018.16.4.e18. Cai T, Liu Y, Xiao J. 2018. Long noncoding RNA MALAT1 knock- Dong N, Guo J, Han S, Bao L, Diao Y, Lin Z. 2019. Positive feed- down reverses chemoresistance to temozolomide via pro- back loop of lncRNA HOXC-AS2/miR-876-5p/ZEB1 to regu- moting microRNA-101 in glioblastoma. Cancer Med. 7 late EMT in glioma. Onco Targets Ther. 12:7601–7609. (4):1404–1415. doi:10.1002/cam4.1384. doi:10.2147/OTT.S216134. Chae Y, Roh J, Kim W. 2021. The roles played by long non- Duan S, Li M, Wang Z, Wang L, Liu Y. 2018. H19 induced by oxi- coding RNAs in glioma resistance. Int J Mol Sci. 22(13) dative stress confers temozolomide resistance in human doi:10.3390/ijms22136834. glioma cells via activating NF-κB signaling. Onco Targets Chang Y-Z, Chai R-C, Pang B, Chang X, SY A, Zhang K-N, Jiang T, Ther. 11:6395–6404. doi:10.2147/OTT.S173244. Wang Y-Z. 2021. METTL3 enhances the stability of MALAT1 Gong R, Li Z-Q, Fu K, Ma C, Wang W, Chen J-C. 2021. Long non- with the assistance of HuR via m6A modification and acti- coding RNA PVT1 promotes stemness and temozolomide vates NF-κB to promote the malignant progression of IDH- resistance through miR-365/ELF4/SOX2 axis in glioma. Exp wildtype glioma. Cancer Lett. 511:36–46. doi:10.1016/j. Neurobiol. 30(3):244–255. doi:10.5607/en20060. canlet.2021.04.020. González-Gómez P, Anselmo NP, Mira H. 2014. BMPs as thera- Chen J, Han P, Dahiya S. 2022a. Glioblastoma: changing con- peutic targets and biomarkers in astrocytic glioma. Biomed cepts in the WHO CNS5 classification. Indian J Pathol Res Int. 2014:549742. doi:10.1155/2014/549742. Microbiol. 65(Supplement):S24–S32. doi:10.4103/ijpm. Gudbjartsson DF, Holm H, Gretarsdottir S, Thorleifsson G, ijpm_1109_21. Walters GB, Thorgeirsson G, Gulcher J, Mathiesen EB, Chen K, Wang X, Wei B, Sun R, Wu C, Yang H. 2022b. LncRNA Njølstad I, Nyrnes A, et al. 2009. A sequence variant in SNHG6 promotes glycolysis reprogramming in hepatocellu- ZFHX3 on 16q22 associates with atrial fibrillation and lar carcinoma by stabilizing the BOP1 protein. Anim Cells ischemic stroke. Nat Genet. 41(8):876–878. doi:10.1038/ng. Syst (Seoul). 26(6):369–379. doi:10.1080/19768354.2022. Gudjonsson T, Altmeyer M, Savic V, Toledo L, Dinant C, Grøfte Chen W, Xu X-K, Li J-L, Kong K-K, Li H, Chen C, He J, Wang F, Li P, M, Bartkova J, Poulsen M, Oka Y, Bekker-Jensen S, et al. 2012. Ge X-S, Li F-C. 2017. MALAT1 is a prognostic factor in glio- TRIP12 and UBR5 suppress spreading of chromatin ubiquity- blastoma multiforme and induces chemoresistance to temo- lation at damaged chromosomes. Cell. 150(4):697–709. zolomide through suppressing miR-203 and promoting doi:10.1016/j.cell.2012.06.039. thymidylate synthase expression. Oncotarget. 8(14):22783– Hausinger RP. 2004. FeII/alpha-ketoglutarate-dependent 22799. doi:10.18632/oncotarget.15199. hydroxylases and related enzymes. Crit Rev Biochem Mol Chen Y, Guo Y, Chen H, Ma F. 2020. Long non-coding RNA Biol. 39(1):21–68. doi:10.1080/10409230490440541. expression profiling identifies a four-long non-coding RNA Hayward D, Cole PA. 2016. LSD1 Histone Demethylase Assays prognostic signature for isocitrate dehydrogenase mutant and Inhibition; Methods Enzymol. 573:261–278. doi:10. glioma. Front Neurol. 11:573264. doi:10.3389/fneur.2020. 1016/bs.mie.2016.01.020. 573264. 26 J. ROH ET AL. He Z, Long J, Yang C, Gong B, Cheng M, Wang Q, Tang J. 2020. Lee SY. 2016. Temozolomide resistance in glioblastoma multi- LncRNA DGCR5 plays a tumor-suppressive role in glioma via forme. Genes Dis. 3(3):198–210. doi:10.1016/j.gendis.2016. the miR-21/Smad7 and miR-23a/PTEN axes. Aging. 12 04.007. (20):20285–20307. doi:10.18632/aging.103800. Leeper HE, Caron AA, Decker PA, Jenkins RB, Lachance DH, Horbinski C, Berger T, Packer RJ, Wen PY. 2022. Clinical impli- Giannini C. 2015. IDH mutation, 1p19q codeletion and cations of the 2021 edition of the WHO classification of ATRX loss in WHO grade II gliomas. Oncotarget. 6 central nervous system tumours. Nat Rev Neurol. 18 (30):30295–30305. doi:10.18632/oncotarget.4497. (9):515–529. doi:10.1038/s41582-022-00679-w. Leighton F, Poole B, Lazarow PB, de Duve C. 1969. The synthesis Huang G-H, Pei Y-C, Yang L, Mou K-J, Tang J-H, Xiang Y, Liu J, Lv and turnover of rat liver peroxisomes. I. fractionation of per- S-Q. 2020. Integrative transcriptome analysis identified a oxisome proteins. J Cell Biol. 41(2):521–535. doi:10.1083/jcb. BMP signaling pathway-regulated lncRNA AC068643.1 in 41.2.521. IDH mutant and wild-type glioblastomas. Oncol Lett. 20 Li H, Yuan X, Yan D, Li D, Guan F, Dong Y, Wang H, Liu X, Yang B. (1):75–84. doi:10.3892/ol.2020.11542. 2017. Long non-coding RNA MALAT1 decreases the sensi- Hurley JH, Dean AM, Koshland DE, Stroud RM. 1991. Catalytic tivity of resistant glioblastoma cell lines to temozolomide. mechanism of NADP(+)-dependent isocitrate dehydrogen- Cell Physiol Biochem. 42(3):1192–1201. doi:10.1159/ ase: implications from the structures of magnesium-isoci- 000478917. trate and NADP+ complexes. Biochemistry. 30(35):8671– Li J, Chen Y, Zhao J, Kong F, Zhang Y. 2011. miR-203 reverses 8678. doi:10.1021/bi00099a026. chemoresistance in p53-mutated colon cancer cells Itsumi M, Inoue S, Elia AJ, Murakami K, Sasaki M, Lind EF, Brenner through downregulation of Akt2 expression. Cancer Lett. D, Harris IS, Chio IIC, Afzal S, et al. 2015. Idh1 protects murine 304(1):52–59. doi:10.1016/j.canlet.2011.02.003. hepatocytes from endotoxin-induced oxidative stress by Li R, Chen W, Mao P, Wang J, Jing J, Sun Q, Wang M, Yu X. 2021a. regulating the intracellular NADP(+)/NADPH ratio. Cell Identification of a three-long non-coding RNA signature for Death Differ. 22(11):1837–1845. doi:10.1038/cdd.2015.38. predicting survival of temozolomide-treated isocitrate dehy- Jiang P, Wang P, Sun X, Yuan Z, Zhan R, Ma X, Li W. 2016. drogenase mutant low-grade gliomas. Exp Biol Med Knockdown of long noncoding RNA H19 sensitizes human (Maywood). 246(2):187–196. doi:10.1177/1535370220962715. glioma cells to temozolomide therapy. Onco Targets Ther. Li Z, Meng X, Wu P, Zha C, Han B, Li L, Sun N, Qi T, Qin J, 9:3501–3509. doi:10.2147/OTT.S96278. Zhang Y, et al. 2021b. Glioblastoma cell-derived lncRNA- Jiang W, Wang S, Xiao M, Lin Y, Zhou L, Lei Q, Xiong Y, Guan K- containing exosomes induce microglia to produce comp- L, Zhao S. 2011. Acetylation regulates gluconeogenesis by lement C5, promoting chemotherapy resistance. Cancer promoting PEPCK1 degradation via recruiting the UBR5 ubi- Immunol Res. 9(12):1383–1399. doi:10.1158/2326-6066.CIR- quitin ligase. Mol Cell. 43(1):33–44. doi:10.1016/j.molcel. 21-0258. 2011.04.028. Lin J, Lin Y, Fan L, Kuang W, Zheng L, Wu J, Shang P, Wang Q, Karreth FA, Tay Y, Perna D, Ala U, Tan SM, Rust AG, DeNicola G, Tan J. 2016. miR-203 inhibits cell proliferation and promotes Webster KA, Weiss D, Perez-Mancera PA, et al. 2011. In vivo cisplatin induced cell death in tongue squamous cancer. identification of tumor- suppressive PTEN ceRNAs in an Biochem Biophys Res Commun. 473(2):382–387. doi:10. oncogenic BRAF-induced mouse model of melanoma. Cell. 1016/j.bbrc.2016.02.105. 147(2):382–395. doi:10.1016/j.cell.2011.09.032. Liu ZZ, Tian YF, Wu H, Ouyang SY, Kuang WL. 2020. LncRNA Katoh M. 2016. Mutation spectra of histone methyltransferases H19 promotes glioma angiogenesis through miR-138/HIF- with canonical SET domains and EZH2-targeted therapy. 1α/VEGF axis. Neoplasma. 67(1):111–118. doi:10.4149/neo_ Epigenomics. 8(2):285–305. doi:10.2217/epi.15.89. 2019_190121N61. Koh H-J, Lee S-M, Son B-G, Lee S-H, Ryoo ZY, Chang K-T, Park J- Loenarz C, Schofield CJ. 2008. Expanding chemical biology of W, Park D-C, Song BJ, Veech RL, et al. 2004. Cytosolic NADP 2-oxoglutarate oxygenases. Nat Chem Biol. 4(3):152–156. +-dependent isocitrate dehydrogenase plays a key role in doi:10.1038/nchembio0308-152. lipid metabolism. J Biol Chem. 279(38):39968–39974. Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella- doi:10.1074/jbc.M402260200. Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues Komori T. 2022. Grading of adult diffuse gliomas according to P, Ellison DW. 2016. The 2016 world health organization the 2021 WHO classification of tumors of the central nervous classification of tumors of the central nervous system: a system. Lab Invest. 102(2):126–133. doi:10.1038/s41374-021- summary. Acta Neuropathol. 131(6):803–820. doi:10.1007/ 00667-6. s00401-016-1545-1. Kuang Y, Bing Z, Jin X, Li Q. 2021. LncRNA H19 upregulation Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella- participates in the response of glioma cells to radiation. Branger D, Hawkins C, Ng HK, Pfister SM, Reifenberger G, Biomed Res Int. 2021:1728352. doi:10.1155/2021/1728352. et al. 2021. The 2021 WHO classification of tumors of the Lee S, Kim YN, Im D, Cho SH, Kim J, Kim J-H, Kim K. 2021. DNA central nervous system: a summary. Neuro Oncol. 23 methylation and gene expression patterns are widely (8):1231–1251. doi:10.1093/neuonc/noab106. altered in fetal growth restriction and associated with FGR Lu C, Ward PS, Kapoor GS, Rohle D, Turcan S, Abdel-Wahab O, development. Anim Cells Syst (Seoul). 25(3):128–135. Edwards CR, Khanin R, Figueroa ME, Melnick A, et al. 2012. doi:10.1080/19768354.2021.1925741. IDH mutation impairs histone demethylation and results in Lee SH, Jo SH, Lee SM, Koh HJ, Song H, Park JW, Lee WH, Huh a block to cell differentiation. Nature. 483(7390):474–478. TL. 2004. Role of NADP+-dependent isocitrate dehydrogen- doi:10.1038/nature10860. ase (NADP+-ICDH) on cellular defence against oxidative Lu C, Wei Y, Wang X, Zhang Z, Yin J, Li W, Chen L, Lyu X, Shi Z, injury by gamma-rays. Int J Radiat Biol. 80(9):635–642. Yan W, You Y. 2020. DNA-methylation-mediated activating doi:10.1080/09553000400007680. of lncRNA SNHG12 promotes temozolomide resistance in ANIMAL CELLS AND SYSTEMS 27 glioblastoma. Mol Cancer. 19(1):28. doi:10.1186/s12943-020- Shao Y, Ye M, Li Q, Sun W, Ye G, Zhang X, Yang Y, Xiao B, Guo J. 1137-5. 2016. LncRNA-RMRP promotes carcinogenesis by acting as a Ludwig K, Kornblum HI. 2017. Molecular markers in glioma. J miR-206 sponge and is used as a novel biomarker for gastric Neurooncol. 134(3):505–512. doi:10.1007/s11060-017-2379-y. cancer. Oncotarget. 7(25):37812–37824. doi:10.18632/ MacDonald MJ, Brown LJ, Longacre MJ, Stoker SW, Kendrick oncotarget.9336. MA, Hasan NM. 2013. Knockdown of both mitochondrial iso- Sun H, Yin L, Li S, Han S, Song G, Liu N, Yan C. 2013. Prognostic citrate dehydrogenase enzymes in pancreatic beta cells inhi- significance of IDH mutation in adult low-grade gliomas: a bits insulin secretion. Biochim Biophys Acta. 1830(11):5104– meta-analysis. J Neurooncol. 113(2):277–284. doi:10.1007/ 5111. doi:10.1016/j.bbagen.2013.07.013. s11060-013-1107-5. Marín-Béjar O, Marchese FP, Athie A, Sánchez Y, González J, Tay Y, Rinn J, Pandolfi PP. 2014. The multilayered complexity of Segura V, Huang L, Moreno I, Navarro A, Monzó M, et al. ceRNA crosstalk and competition. Nature. 505(7483):344– 2013. Pint lincRNA connects the p53 pathway with epige- 352. doi:10.1038/nature12986. netic silencing by the polycomb repressive complex 2. Tian T, Mingyi M, Qiu X, Qiu Y. 2016. MicroRNA-101 reverses Genome Biol. 14(9):R104. doi:10.1186/gb-2013-14-9-r104. temozolomide resistance by inhibition of GSK3β in glioblas- McNamara MG, Sahebjam S, Mason WP. 2013. Emerging bio- toma. Oncotarget. 7(48):79584–79595. doi:10.18632/ markers in glioblastoma. Cancers (Basel). 5(3):1103–1119. oncotarget.12861. doi:10.3390/cancers5031103. Turcan S, Rohle D, Goenka A, Walsh LA, Fang F, Yilmaz E, Mercer TR, Dinger ME, Mattick JS. 2009. Long non-coding RNAs: Campos C, Fabius AWM, Lu C, Ward PS, et al. 2012. IDH1 insights into functions. Nat Rev Genet. 10(3):155–159. doi:10. mutation is sufficient to establish the glioma hypermethyla- 1038/nrg2521. tor phenotype. Nature. 483(7390):479–483. doi:10.1038/ Nobusawa S, Watanabe T, Kleihues P, Ohgaki H. 2009. IDH1 nature10866. mutations as molecular signature and predictive factor of Waitkus MS, Diplas BH, Yan H. 2018. Biological role and thera- secondary glioblastomas. Clin Cancer Res. 15(19):6002– peutic potential of IDH mutations in cancer. Cancer Cell. 34 6007. doi:10.1158/1078-0432.CCR-09-0715. (2):186–195. doi:10.1016/j.ccell.2018.04.011. Peng Z, Liu C, Wu M. 2018. New insights into long noncoding Ward PS, Patel J, Wise DR, Abdel-Wahab O, Bennett BD, Coller RNAs and their roles in glioma. Mol Cancer. 17(1):61. doi:10. HA, Cross JR, Fantin VR, Hedvat CV, Perl AE, et al. 2010. The 1186/s12943-018-0812-2. common feature of leukemia-associated IDH1 and IDH2 Pyko IV, Nakada M, Sabit H, Teng L, Furuyama N, Hayashi Y, mutations is a neomorphic enzyme activity converting Kawakami K, Minamoto T, Fedulau AS, Hamada J. 2013. alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell. 17 Glycogen synthase kinase 3β inhibition sensitizes human (3):225–234. doi:10.1016/j.ccr.2010.01.020. glioblastoma cells to temozolomide by affecting O6-methyl- Weller M, Wick W, Aldape K, Brada M, Berger M, Pfister SM, guanine DNA methyltransferase promoter methylation via Nishikawa R, Rosenthal M, Wen PY, Stupp R, Reifenberger c-Myc signaling. Carcinogenesis. 34(10):2206–2217. doi:10. G. 2015. Glioma. Nat Rev Dis Primers. 1:15017. doi:10.1038/ 1093/carcin/bgt182. nrdp.2015.17. Rahman Z, Bazaz Mohd R, Devabattula G, Khan Mohd A, Wick W, Hartmann C, Engel C, Stoffels M, Felsberg J, Godugu C. 2021. Targeting H3K9 methyltransferase G9a Stockhammer F, Sabel MC, Koeppen S, Ketter R, and its related molecule GLP as a potential therapeutic strat- Meyermann R, et al. 2009. NOA-04 randomized phase III egy for cancer. J Biochem Mol Toxicol. 35(3). doi:10.1002/jbt. trial of sequential radiochemotherapy of anaplastic glioma 22674. with procarbazine, lomustine, and vincristine or temozolo- Reitman ZJ, Yan H. 2010. Isocitrate dehydrogenase 1 and 2 mide. J Clin Oncol. 27(35):5874–5880. doi:10.1200/JCO. mutations in cancer: alterations at a crossroads of cellular 2009.23.6497. metabolism. J Natl Cancer Inst. 102(13):932–941. doi:10. Wu F, Zhao Z, Chai R, Liu Y, Wang K, Wang Z, Li G, Huang R, 1093/jnci/djq187. Jiang H, Zhang K. 2018. Expression profile analysis of anti- Rickert DE. 1987. Metabolism of nitroaromatic compounds. Drug sense long non-coding RNA identifies WDFY3-AS2 as a prog- Metab Rev. 18(1):23–53. doi:10.3109/03602538708998299. nostic biomarker in diffuse glioma. Cancer Cell Int. 18:107. Sachdeva R, Wu M, Johnson K, Kim H, Celebre A, Shahzad U, doi:10.1186/s12935-018-0603-2. Graham MS, Kessler JA, Chuang JH, Karamchandani J, et al. Xiong Z, Wang L, Wang Q, Yuan Y. 2018. LncRNA MALAT1/ 2019. BMP signaling mediates glioma stem cell quiescence miR-129 axis promotes glioma tumorigenesis by targeting and confers treatment resistance in glioblastoma. Sci Rep. SOX2. J Cell Mol Med. 22(8):3929–3940. doi:10.1111/jcmm. 9(1):14569. doi:10.1038/s41598-019-51270-1. 13667. Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP. 2011. A ceRNA Xu W, Yang H, Liu Y, Yang Y, Ping W, Kim S-H, Ito S, Yang C, Pu hypothesis: the rosetta stone of a hidden RNA language? W, Xiao M-T, et al. 2011. Oncometabolite 2-hydroxyglutarate Cell. 146(3):353–358. doi:10.1016/j.cell.2011.07.014. is a competitive inhibitor of α-ketoglutarate-dependent Seo D, Kim D, Chae Y, Kim W. 2020. The ceRNA network of dioxygenases. Cancer Cell. 19(1):17–30. doi:10.1016/j.ccr. lncRNA and miRNA in lung cancer. Genomics Inform. 18(4): 2010.12.014. e36. doi:10.5808/GI.2020.18.4.e36. Xu X, Zhao J, Xu Z, Peng B, Huang Q, Arnold E, Ding J. 2004. Seo D, Kim D, Kim W. 2019. Long non-coding RNA linc00152 Structures of human cytosolic NADP-dependent isocitrate acting as a promising oncogene in cancer progression. dehydrogenase reveal a novel self-regulatory mechanism Genomics Inform. 17(4):e36. doi:10.5808/GI.2019.17.4.e36. of activity. J Biol Chem. 279(32):33946–33957. doi:10.1074/ Seo D, Roh J, Chae Y, Kim W. 2021. Gene expression profiling jbc.M404298200. after LINC00472 overexpression in an NSCLC cell line1. Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan W, Cancer Biomark. 32(2):175–188. doi:10.3233/CBM-210242. Kos I, Batinic-Haberle I, Jones S, Riggins GJ, et al. 2009. 28 J. ROH ET AL. IDH1 and IDH2 mutations in gliomas. N Engl J Med. 360 Zhang Y, Zhou S, Yan H, Xu D, Chen H, Xiao-yan W, Xiao W, Liu (8):765–773. doi:10.1056/NEJMoa0808710. Y, Zhang L, Wang S, et al. 2016. miR-203 inhibits proliferation Yang B, Zhong C, Peng Y, Lai Z, Ding J. 2010. Molecular mech- and self-renewal of leukemia stem cells by targeting survivin anisms of “off-on switch” of activities of human IDH1 by and Bmi-1. Sci Rep. 6:19995. doi:10.1038/srep19995. tumor-associated mutation R132H. Cell Res. 20(11):1188– Zhang Z, Yin J, Lu C, Wei Y, Zeng A, You Y. 2019b. Exosomal 1200. doi:10.1038/cr.2010.145. transfer of long non-coding RNA SBF2-AS1 enhances che- Youssef G, Miller JJ. 2020. Lower grade gliomas. Curr moresistance to temozolomide in glioblastoma. J Exp Clin Neurol Neurosci Rep. 20(7):21. doi:10.1007/s11910-020- Cancer Res. 38(1):166. doi:10.1186/s13046-019-1139-6. 01040-8. Zhao S, Lin Y, Xu W, Jiang W, Zha Z, Wang P, Yu W, Li Z, Gong L, Yu H, Zheng J, Liu X, Xue Y, Shen S, Zhao L, Li Z, Liu Y. 2017. Peng Y, et al. 2009. Glioma-derived mutations in IDH1 dom- Transcription factor NFAT5 promotes glioblastoma cell- inantly inhibit IDH1 catalytic activity and induce HIF-1alpha. driven angiogenesis via SBF2-AS1/miR-338-3p-mediated Science. 324(5924):261–265. doi:10.1126/science.1170944. EGFL7 expression change. Front Mol Neurosci. 10:301. Zhou Q, Liu Z-Z, Wu H, Kuang W-L. 2022. LncRNA H19 pro- doi:10.3389/fnmol.2017.00301. motes cell proliferation, migration, and angiogenesis of Zhang J, Zhang P, Wang L, Piao H, Ma L. 2014. Long non-coding glioma by regulating Wnt5a/β-catenin pathway via target- RNA HOTAIR in carcinogenesis and metastasis. Acta Biochim ing miR-342. Cell Mol Neurobiol. 42(4):1065–1077. doi:10. Biophys Sin (Shanghai). 46(1):1–5. doi:10.1093/abbs/ 1007/s10571-020-00995-z. gmt117. Zhou Y, Hu X-W, Yang S-J, Yu Z. 2020. Knockdown of Zhang Q, Liu X-J, Li Y, Ying X-W, Chen L. 2021. Prognostic value lncRNAZFAS1 suppresses cell proliferation and metastasis of immune-related lncRNA SBF2-AS1 in diffuse lower-grade in non-small cell lung cancer. Anim Cells Syst (Seoul). 24 glioma. Technol Cancer Res Treat. 20:15330338211011966. (2):107–113. doi:10.1080/19768354.2020.1736623. doi:10.1177/15330338211011966. Zou H, Wu L-X, Yang Y, Li S, Mei Y, Liu Y-B, Zhang L, Cheng Y, Zhang Y, Yang G, Luo Y. 2019a. Long non-coding RNA PVT1 Zhou H-H. 2017. lncRNAs PVT1 and HAR1A are prognosis promotes glioma cell proliferation and invasion by targeting biomarkers and indicate therapy outcome for diffuse miR-200a. Exp Ther Med. 17(2):1337–1345. doi:10.3892/etm. glioma patients. Oncotarget. 8(45):78767–78780. doi:10. 2018.7083. 18632/oncotarget.20226. http://www.deepdyve.com/assets/images/DeepDyve-Logo-lg.png Animal Cells and Systems Taylor & Francis

Long non-coding RNA in glioma: novel genetic players in temozolomide resistance

Loading next page...
 
/lp/taylor-francis/long-non-coding-rna-in-glioma-novel-genetic-players-in-temozolomide-yLWigVxeK0

References (98)

Publisher
Taylor & Francis
Copyright
© 2023 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group
ISSN
1976-8354
eISSN
2151-2485
DOI
10.1080/19768354.2023.2175497
Publisher site
See Article on Publisher Site

Abstract

ANIMAL CELLS AND SYSTEMS 2023, VOL. 27, NO. 1, 19–28 https://doi.org/10.1080/19768354.2023.2175497 Long non-coding RNA in glioma: novel genetic players in temozolomide resistance a# a# b c a,d Jungwook Roh , Mijung Im , JiHoon Kang , BuHyun Youn and Wanyeon Kim a b Department of Science Education, Korea National University of Education, Cheongju-si, Republic of Korea; Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory, Emory University School of Medicine, Atlanta, GA, USA; Department of Biological Sciences, Pusan National University, Busan, Republic of Korea; Department of Biology Education, Korea National University of Education, Cheongju-si, Republic of Korea ABSTRACT ARTICLE HISTORY Received 25 November 2022 Glioma is the most common primary malignant brain tumor in adults and accounts for Revised 15 January 2023 approximately 80% of brain and central nervous system tumors. In 2021, the World Health Accepted 27 January 2023 Organization (WHO) published a new taxonomy for glioma based on its histological features and molecular alterations. Isocitrate dehydrogenase (IDH) catalyzes the decarboxylation of KEYWORDS isocitrate, a critical metabolic reaction in energy generation in cells. Mutations in the IDH genes Isocitrate dehydrogenase; interrupt cell differentiation and serve as molecular biomarkers that can be used to classify long non-coding RNA; gliomas. For example, the mutant IDH is widely detected in low-grade gliomas, whereas the glioma; temozolomide wild type is in high-grade ones, including glioblastomas. Long non-coding RNAs (lncRNAs) are resistance epigenetically involved in gene expression and contribute to glioma development. To investigate the potential use of lncRNAs as biomarkers, we examined lncRNA dysregulation dependent on the IDH mutation status. We found that several lncRNAs, namely, AL606760.2, H19, MALAT1, PVT1 and SBF2-AS1 may function as glioma risk factors, whereas AC068643.1, AC079228.1, DGCR5, FAM13A-AS1, HAR1A and WDFY3-AS2 may have protective effects. Notably, H19, MALAT1, PVT1, and SBF2-AS1 have been associated with temozolomide resistance in glioma patients. This review study suggests that targeting glioma-associated lncRNAs might aid the treatment of glioma. Introduction reclassifies gliomas into adult-type diffuse gliomas, pedi- Gliomas are primary brain tumors derived from neuro- atric-type diffuse LGGs, pediatric-type diffuse HGGs, cir- glial stem cells or neuroglial progenitor cells (Weller cumscribed astrocytic gliomas, and ependymal tumors et al. 2015). The grading of gliomas based on histological (Horbinski et al. 2022). This system also classifies features has been debated for decades by technical gliomas according to isocitrate dehydrogenase (IDH) limitations and different opinions. However, recent mutation status and uses various molecular markers to advances in molecular genetics have revealed novel classify subgroups by their molecular characteristics changes in several tumors and have changed the diag- (Komori 2022). nostic paradigm for tumors of the central nervous IDH is an essential enzyme involved in major meta- system (CNS). Molecular diagnosis was first introduced bolic processes such as the TCA cycle, glutamine in the World Health Organization (WHO) CNS glioma metabolism, adipogenesis, and redox regulation and classification system published in 2016 (Louis et al. has three isoforms, and IDH1 and IDH2 are used as 2016; Louis et al. 2021). According to this system, glioma biomarkers (Koh et al. 2004; Lee et al. 2004; grades 1 and 2 are low-glioma grades (LGGs), and Yan et al. 2009). The most common mutation detected grades 3 and 4 are high-glioma grades (HGGs) (Chen in human IDH1 replaces arginine at position 132 by et al. 2022). In addition, a new classification in the histidine (R132H), whereas the one in human IDH2 WHO published in 2021 incorporates more molecular does arginine at 172 by lysine (R172 K) (Dang et al. alterations into the diagnosis of many tumors and 2009; Yan et al. 2009). Wildtype IDH enzymes convert CONTACT BuHyun Youn bhyoun72@pusan.ac.kr Department of Biological Sciences, Pusan National University, Busandaehak-ro 63beon-gil 2, Geumjeong-gu, Busan 46241, Republic of Korea; Wanyeon Kim wykim82@knue.ac.kr Department of Biology Education, Korea National University of Education, 250 Taeseongtabyeon-ro, Gangnae-myeon, Heungdeok-gu, Cheongju-si, Chungbuk 28173, Republic of Korea Authors contributed equally. © 2023 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. 20 J. ROH ET AL. isocitrate to α-ketoglutarate (α-KG). However, the Li et al. 2021). In this review, to address the issue of mutant IDHs have changed their substrate specificity, TMZ resistance, we summarize the effects of lncRNA converting α-KG to 2-hydroxyglutarate (2-HG). The con- dysregulation according to IDH mutation status in sequent excessive accumulation of 2-HG leads to glioma. glioma development and their malignant progression, associated metabolic alterations, DNA methylation, IDHs as classification biomarkers in glioma and reactive oxygen species (ROS) production (Dang et al. 2009; Lu et al. 2012; Turcan et al. 2012). The IDH is a representative biomarker of glioma classifi- majority of LGG patients have mutations in IDH, and cation, and WHO primarily classifies gliomas according a significant correlation has been reported between to IDH mutation status. IDH is an enzyme that catalyzes IDH mutation and improved overall survival in LGG the reversible oxidative decarboxylation of isocitrate, (Rickert 1987; Youssef and Miller 2020). WHO CNS which results in the production of carbon dioxide and grade 4 glioblastoma can be classified as primary or α-KG in the TCA cycle during glucose metabolism, secondary. Most primary and secondary glioblastomas which in turn, is coupled with the transformation of are of the IDH-wildtype or mutant types, respectively, NAD(P) to NAD(P)H (Zhao et al. 2009). In mammals, and overall survival is better for the secondary type IDH exists in three isoforms: IDH1 is present in the cyto- (Nobusawa et al. 2009; Wick et al. 2009; Sun et al. plasm and peroxisomes, and IDH2 and IDH3, in the mito- 2013). However, diagnoses of glioma based on the chondrial matrix (Leighton et al. 1969). IDH1 and IDH2 mutation status of the IDH genes alone overlook the use NADP as a cofactor to produce α-KG and NADPH, various origins and characteristics of glioma. Therefore, and the generated NADPH protects cells from oxidative an understanding of the molecular mechanism of damage by participating in key ROS scavenging pro- glioma is required for accurate diagnosis and cesses such as biosynthetic reactions and glutathione treatment. disulfide reduction (Hurley et al. 1991; Xu et al. 2004; Long non-coding RNAs (lncRNAs), which do not Itsumi et al. 2015). IDH3 plays a central role in energy encode proteins and are more than 200 nucleotides in production in the TCA cycle by generating α-KG and length, regulate cellular biological processes in cancer NADH using NAD as a cofactor (Reitman and Yan such as cell proliferation, metastasis, invasion, motility, 2010). Both IDH1 and IDH2 form a homodimer, and drug resistance, and their intracellular locations whereas IDH3 holoenzyme is multimeric and assembled reflect their functions (Seo et al. 2019; Seo et al. 2021; with multiple subunits of α, β, and γ encoded by IDH3a, Chen et al. 2022). Nuclear lncRNAs affect gene IDH3b, and IDH3c, respectively (MacDonald et al. 2013). expression at the transcriptional level, regulating In diffuse gliomas, the IDH1-mutant type is the most histone modification, DNA methylation and chromatin common and observed in more than 90% of cases. The remodeling (Marín-Béjar et al. 2013; Zhang et al. 2014; IDH2-mutant type is observed in approximately 10% of Chu et al. 2015; Do et al. 2019). On the other hand, cyto- cases, but the IDH3-mutant type has never been plasmic lncRNAs are competitive endogenous RNAs observed (Waitkus et al. 2018). (ceRNAs) that interact with mRNAs and miRNAs to epi- In cases of IDH1 mutation, the homodimer formed by genetically regulate gene expression at the post-tran- two IDH1-mutant monomers does not have enzymatic scriptional and translational levels. Recent studies activity, but the heterodimer composed of the IDH-wild- suggest that dysregulations of lncRNA can result in the type and mutant monomer has enzymatic activity initiation, progression, and other malignant phenotypes sufficient to convert α-KG to 2-HG (Dang et al. 2009; of glioma (Karreth et al. 2011; Tay et al. 2014; Shao et al. Yang et al. 2010). IDH is one of the most studied genes 2016; Peng et al. 2018; Zhou et al. 2020). in glioma development, and cancers with IDH1 or IDH2 O -methylguanine-DNA methyltransferase (MGMT) mutations produce 10–100 times more 2-HG than methylation, 1p/19q co-deletion, IDH, and epidermal those with the IDH1 or IDH2 wildtype (Ward et al. growth factor receptor have been widely used as 2010; Leeper et al. 2015). IDH1 mutation converts α-KG glioma biomarkers (McNamara et al. 2013;Ludwig to 2-HG, which accumulates in the cytoplasm, and and Kornblum 2017). Recently, it has been demon- thereby releases carbon from the TCA cycle. This cycle strated that lncRNAs might also function as molecular utilizes metabolic reprogramming to compensate for biomarkers in glioma (Peng et al. 2018). For example, fluctuations in metabolic pathways; for example, gluta- lncRNA dysregulation has been shown to be corre- mate dehydrogenase converts glutamine to α-KG, lated with glioma malignancy and associated with which replenishes the TCA cycle (Hausinger 2004; resistance to radiotherapy and temozolomide (TMZ) Loenarz and Schofield 2008; Xu et al. 2011). In addition (Lu et al. 2020;Chae etal. 2021;Kuang et al. 2021; to the metabolic changes, the hypermethylation of ANIMAL CELLS AND SYSTEMS 21 CpG islands is also affected by IDH mutations. The tumor behavior and prognosis. LncRNA AC068643.1 is glioma-specific methylation pattern provides clues more highly expressed in IDH-mutant glioblastomas regarding the pathogenesis of IDH-mutant gliomas. than in the wildtype. The expression levels of DNA methylation is regulated by methyltransferase AC068643.1 are positively correlated with the protein and demethylase. During demethylation, ten-eleven levels of bone morphogenetic protein (BMP) and myos- translocation methylcytosine dioxygenase (TET) con- tatin (MSTN), and it has been experimentally demon- verts 5-methylcytosine to cytosine in an iron- and α- strated that both BMP and MSTN directly stimulate KG-dependent manner. Furthermore, the activity of AC068643.1 expression (Huang et al. 2020). BMP inhibits TET might be inhibited by 2-HG due to its structural simi- cell proliferation in glioblastoma and astrocytic glioma, larity to α-KG (Xu et al. 2011; Lee et al. 2021). Histone and a BMP-like synthetic is considered a non-cytotoxic methylation status is regulated by histone methyltrans- therapeutic that might prevent glioma growth and ferases such as euchromatic histone lysine methyltrans- recurrence. Altogether, these results suggest that ferase 1 (EHMT1), EHMT2, SET nuclear proto-oncogene AC068643.1 can function as a protective lncRNA in glio- (SET), and enhancer of zeste 2 polycomb repressive blastoma patients (González-Gómez et al. 2014; Sach- complex 2 subunit (EZH2), and demethylases such as deva et al. 2019). lysine demethylase 1A (KDM1A) and KDM4A (Chowdh- Differentially expressed lncRNAs were identified ury et al. 2011; Hayward and Cole 2016; Katoh 2016; using GSE107850, a dataset of glioma patients that Rahman et al. 2021). Like TET, histone demethylases underwent surgical excision and TMZ treatment. A such as KDM4 and KDM5 are inhibited by high levels risky lncRNA AL606760.2 and two protective lncRNAs, of 2-HG (Chowdhury et al. 2011). Many studies report FAM13A antisense RNA 1 (FAM13A-AS1) and that histone methylation markers accumulate in AC079228.1, were suggested as predictors of TMZ various cancers harboring a mutant IDH gene. Also, inhi- efficacy in IDH-mutant type LGGs (Li et al. 2021). Further- bition of histone demethylation by 2-HG leads to more, levels of AL606760.2 and Smad2, which mediates impaired cell differentiation, which might be associated transforming growth factor (TGF)-beta signaling, are with the tumorigenesis of IDH-mutant glioma (Lu et al. positively correlated. Thus, AL606760.2 appears to regu- 2012). late intracellular processes such as cell proliferation, IDH is an essential enzyme for energy metabolism apoptosis, and differentiation (Abdel-Wahab et al. associated with the TCA cycle, and mutated IDH causes 2002). On the other hand, FAM13A-AS1 is involved in 2-HG accumulation, hypermethylation, and oxidative decreasing cell proliferation and differentiation, and its damage. Because of these alterations, it is reasonable levels were found to be significantly correlated with to classify glioblastomas by IDH mutational status. The UBR5 levels (an E3 ubiquitin-protein ligase) (Jiang et al. new molecular markers including IDH will help diagnose 2011; Gudjonsson et al. 2012). The two cancer-related and effectively treat gliomas. genes, pre-mRNA processing factor 40 homolog B (PRPF40B) and zinc finger homeobox 3 (ZFHX3), were suggested to be potential target mRNAs of lncRNA LncRNAs and IDH mutations in glioma AC079228.1. PRPF40B is a splicing gene that directly In brain tumors, IDH mutation status is known to be interacts with splicing factor 1 (SF1), and associated associated with a patient’s prognosis of brain tumors with U2 small nuclear RNA auxiliary factor 1 (U2AF1). and with a well-known molecular biomarker of glioma ZFHX3 acts as a transcription factor for regulation of (Turcan et al. 2012). Reportedly, lncRNAs are epigeneti- myogenic and neuronal differentiation and inhibition cally involved in gene expression regulation and treat- of cell cycle progression (Benjamin et al. 2009; Gudb- ment resistance in brain tumors (Mercer et al. 2009). jartsson et al. 2009). Furthermore, accumulating evidence suggests that In another study, lncRNAs differentially expressed in lncRNAs function as ceRNAs that regulate gene IDH-mutant and IDH-wildtype glioma samples obtained expression and alter cell biological properties such as from several CCGA, TCGA, and GSE16011 datasets were cell viability, proliferation, motility, and invasion (Do analyzed using the LNCipedia database (Chen et al. and Kim 2018; Seo et al. 2020). 2020). Multivariate Cox regression analysis of the prog- nostic performances of differentially expressed lncRNAs identified H19 and plasmacytoma variant translocation Dysfunction of lncRNAs according to IDH- 1 (PVT1) as risky lncRNAs and highly accelerated region mutant status 1A (HAR1A) as a protective lncRNA in glioma. LncRNA Glioblastoma can be classified as IDH-wildtype or H19 promotes angiogenesis in gliomas by increasing mutant types, each of which is associated with a distinct the expressions of hypoxia inducible factor 1 subunit 22 J. ROH ET AL. alpha (HIF-1α) and vascular endothelial growth factor highly expressed in IDH-wildtype than in IDH-mutant (VEGF) through targeting miR-138. In glioma, H19 can glioma and its knockdown in glioblastoma upregulates also target miR-342 and upregulate the Wnt5a/β- miR-338-3p, but downregulates EGF-like domain mul- catenin pathway to promote cell proliferation and tiple 7 (EGFL7), thereby inhibiting angiogenesis migration (Liu et al. 2020; Zhou et al. 2022). Also, (Cancer Genome Atlas Research Network et al. 2015; lncRNA PVT1 targets miR-200a to regulate the cell Yu et al. 2017; Zhang et al. 2021). cycle and promote cell proliferation and invasion An analysis of the CGCA database revealed lncRNA (Zhang et al. 2019). On the other hand, lncRNA HAR1A WDFY3 antisense RNA 2 (WDFY3-AS2) is expressed at acts to suppress the progression of diffuse glioma and low levels in IDH-wildtype gliomas. Kaplan-Meier analy- is expressed at low levels in glioma. In addition, glioma sis showed that WDFY3-AS2 overexpression was associ- patients with high PVT1 expression or low HAR1A ated with longer overall survival than low WDFY3-AS2 expression is reported to be prognostic of poor survival expression, and Cox regression analysis showed its in glioma patients. Moreover, the down-regulation of expression was independently correlated with overall PVT1 and up-regulation of HAR1A improved the survival survival. In addition, gene ontology (GO) and gene set of glioma patients that received chemotherapy and enrichment analysis (GSEA) revealed that WDFY3-AS2 radiotherapy. These findings imply that these lncRNAs is involved in the regulation of synaptic transduction, play critical roles in diffuse glioma progression and glutamate receptors, and TNF signaling pathways (Wu that PVT1 and HAR1A should be explored as promising et al. 2018). biomarkers for diagnosis, prognosis, and targeted LncRNA DiGeorge syndrome critical region gene 5 therapy (Zou et al. 2017). (DGCR5) plays cancer-dependent roles. For example, although DGCR5 levels were unrelated to WHO malig- nancy grade in glioma, they were more downregulated LncRNA dysfunction in IDH-wildtype gliomas in IDH-wildtype glioma than in IDH-mutant glioma. Fur- IDH-wildtype gliomas are associated with poor prog- thermore, DGCR5 increased the levels of Smad7 and noses and high CNS grades. Metastasis-associated lung phosphatase and tensin homolog (PTEN) by sponging adenocarcinoma transcript 1 (MALAT1) is an overex- miR-21 and miR-23a, respectively, in glioma cells. It is pressed lncRNA in glioblastoma and promotes cell also reported that Smad7 inhibits the migratory and growth and tumorigenesis. For example, it has been invasive activities of glioma, and PTEN inhibits cell pro- demonstrated that lncRNA MALAT1 promotes the liferation and enhances apoptosis. Patients with low expression of SRY-box transcription factor 2 (SOX2) by DGCR5 expression levels show poorer overall survival suppressing miR-129 to promote glioma stem cell viabi- (He et al. 2020). lity, proliferative abilities, and tumorigenesis (Xiong et al. 2018). In addition, methyltransferase 3 (METTL3), which TMZ resistance by lncRNAs according to IDH upregulates the expression of MALAT1 through N - mutation status methyladenosine (m A) modification, is also associated with poor prognosis. It was also demonstrated that Glioma classification based on histological features and MALAT1 overexpression promotes the malignant IDH mutation status helps with diagnosis and prognosis. glioma phenotype by inducing epithelial–mesenchymal Although advances in techniques, technologies, surgical transition (EMT) and tumor necrosis factor (TNF) signal- resection, chemotherapy, and radiotherapy have signifi- ing through nuclear factor kappa B (NF-κB) activation cantly improved treatment efficacy outcomes fall far (Chang et al. 2021). short of satisfactory. Furthermore, accumulating evi- Furthermore, lncRNA SBF2 antisense RNA 1 (SBF2- dence indicates lncRNA dysregulations are involved in AS1), which plays an oncogenic role in various tumors, glioma chemo- and radioresistance. This review com- may function as a biomarker in diffuse LGG. Kaplan- piles several differentially expressed lncRNAs in IDH- Meier analysis showed that the prognosis of LGG mutant and IDH-wildtype gliomas, and it has been patients with high SBF2-AS1 expression was poorer shown that lncRNAs SBF2-AS1, PVT1, MALAT1, and H19 than those with low expression. Cox regression analysis significantly contribute to the induction of TMZ showed that SBF2-AS1 is an independent prognostic resistance. factor of poorer overall survival in LGG (Zhang et al. TMZ is a lipophilic anticancer drug, which is widely 2021). Most LGGs are IDH mutant type, and only some used to treat brain tumors. TMZ can be readily delivered are IDH-wildtype. However, the molecular and clinical to brain tissues as it crosses the blood–brain barrier and characteristics of IDH-wildtype LGG are similar to those induces cell death by causing single- or double-strand of glioblastoma. It is reported that SBF2-AS1 is more breaks by incorporating mismatched base pairs. ANIMAL CELLS AND SYSTEMS 23 However, more than 50% of glioma patients do not reportedly, its knockdown effectively promotes glioma respond to TMZ, and lncRNAs appear to be involved in cell sensitivity to TMZ. PVT1 and miR-365 expressions TMZ resistance by regulating chemoresistance-associ- are negatively correlated, and the overexpression of ated gene expression, enhancing cell survival, and inhi- miR-365 suppresses TMZ resistance in glioma cells. biting apoptosis (Lee 2016). Furthermore, numerous PVT1 competitively binds to miR-365 and positively studies have suggested that lncRNA might act as a regulates the expression of E74 like ETS transcription ceRNA of miRNA to modulate the expressions of target factor 4 (ELF4), which contributes to the induction of genes (Salmena et al. 2011). The interaction between glioma stemness and TMZ resistance (Bazzoli et al. lncRNA and miRNA might also influence TMZ resistance 2012; Gong et al. 2021). MALAT1 is frequently overex- in glioma cells. It is reported that sponging of miR-151a- pressed in IDH-wildtype gliomas and enhances resist- 3p by lncRNA SBF2-AS1, which is more highly expressed ance to TMZ administration by regulating the in IDH-wildtype than IDH-mutant gliomas, is responsible expressions of miRNAs. Several studies have shown for TMZ resistance in glioma cells (Zhang et al. 2019). that miR-203 contributes to reduced chemoresistance Furthermore the transcription factor, zinc finger E-box in various cancers (Li et al. 2011; Lin et al. 2016; Zhang binding homeobox 1 (ZEB1), might increase SBF2-AS1 et al. 2016). Furthermore, MALAT1 promotes glioma expression by binding to its promotor region, and the cell proliferation and TMZ resistance by decreasing resulting high level of SBF2-AS1 downregulates miR- miR-203 expression and increasing the expression of 151a-3p and the subsequent upregulation of X-ray thymidylate synthase (TS), a potent cancer chemother- repair cross complementing 4 (XRCC4), a target of miR- apy target (Chu et al. 2003; Chen et al. 2017). In addition, 151a-3p. XRCC4 promotes DNA double-strand break MALAT1 downregulates miR-101, consequently increas- repair and thus enhances cell survival and inhibits apop- ing resistance to TMZ in glioma cells by increasing glyco- tosis in glioma cells in response to TMZ treatment. Fur- gen synthase kinase 3β (GSK-3β) levels. Also, increased thermore, it has been shown that SBF2-AS1 depletion GSK-3β expression might be associated with resistance reduces the level of XRCC4, delays the repair of TMZ- to TMZ and poor prognosis in glioma patients by induced DNA damage, and increases glioma cell sensi- decreasing MGMT promoter methylation and thus upre- tivity to TMZ. PVT1 is a known oncogene in various gulation of MGMT (Pyko et al. 2013; Tian et al. 2016; Cai cancers, including glioma. It is expressed at higher et al. 2018). MALAT1 also regulates TMZ resistance by levels in IDH-mutant than in IDH-wildtype gliomas and modulating multidrug resistance-related genes. The Figure 1. Mechanisms of lncRNAs inducing TMZ resistance. H19 is involved in induction of glioma cell viability and survival through upregulation of MDR1, MRP1, and ABCG2 and activation of NF-κB signaling, responsible for induction of TMZ resistance (Jiang et al. 2016; Duan et al. 2018). MALAT1 is associated with glioma cell proliferation, viability, and invasion through regulation of ceRNA net- works with miRNAs, upregulation of multidrug resistance-related genes, and regulation of ZEB1, which might contribute to glioma TMZ resistance (Chu et al. 2003; de Cremoux et al. 2007; Pyko et al. 2013; Tian et al. 2016; Chen et al. 2017; Li et al. 2017; Cai et al. 2018; Dong et al. 2019). PVT1 and SBF2-AS1 play oncogenic roles in induction of glioma stemness and cell survival, respectively, through regulation of ceRNA networks, resulting in TMZ resistance in glioma cells (Bazzoli et al. 2012; Zhang et al. 2019; Gong et al. 2021). 24 J. ROH ET AL. Figure 2. Therapeutic strategies targeting lncRNAs. Downregulation of risky lncRNAs such as AL606760.2 (Li et al. 2021), H19 (Chen et al. 2020), PVT1 (Chen et al. 2020), MALAT1 (Xiong et al. 2018), and SBF2-AS1 (Zhang et al. 2021) and upregulation of protective lncRNAs such as AC068643.1 (Huang et al. 2020), AC079228.1 (Li et al. 2021), FAM13A-AS1 (Li et al. 2021), HAR1A (Chen et al. 2020), DGCR5 (He et al. 2020), and WDFY3-AS2 (Wu et al. 2018) might help predict prognosis in glioma. downregulation of MALAT1 reduces the mRNA plus drug treatment. However, the discovery of expression levels of multidrug resistance-related pro- lncRNAs, known to induce TMZ resistance, does not teins such as multidrug resistance protein 1 (MDR1), completely explain the difficulties of treating patients multidrug resistance protein 5 (MRP5), and lung–resist- with TMZ-resistant glioma. Nonetheless, targeting ance related protein 1 (LRP1), which are important regu- these lncRNAs is a promising way of reducing TMZ lators of chemoresistance, and also reduces glioma cell resistance. sensitivity to TMZ. In addition, MALAT1 is involved in EMT induction and thus regulates glioblastoma cell sen- Conclusions sitivity to TMZ. The downregulation of MALAT1 decreases TMZ resistance by reducing the expression The dysregulation of lncRNA contributes to the develop- level of ZEB1, an EMT-related protein that increases ment of several types of cancer, and lncRNA promotes cancer cell motility and invasiveness (de Cremoux et al. glioma development and induces chemo- and radiore- 2007; Li et al. 2017; Dong et al. 2019). H19 is highly sistance. IDH mutation status is a widely-accepted bio- expressed in IDH-mutant gliomas and involved in TMZ marker for glioma classification, but additional markers resistance by regulating multidrug resistance genes, are desired to determine effective treatment strategies. and its levels are elevated in TMZ-resistant glioma This review focuses on risky and protective lncRNAs cells. Inhibition of H19 downregulates the expressions according to IDH mutation status and the mechanisms of several multidrug resistance genes such as MDR1, involved in TMZ resistance. We suggest five lncRNAs MRP1, and ATP-binding cassette super-family G acting as risk factors, namely, AL606760.2, H19, member 2 (ABCG2) in TMZ-resistant cells at the mRNA MALAT1, PVT1, and SBF2-AS1, and another six protective and protein levels, which suggests H19 plays an impor- lncRNAs, that are AC068643.1, AC079228.1, DGCR5, tant role in the induction of TMZ resistance in glioma FAM13A-AS1, HAR1A, and WDFY3-AS2. The risky H19, cells (Jiang et al. 2016). Furthermore, oxidative stress MALAT1, PVT1, and SBF2-AS1 increase TMZ resistance induces H19 expression and thus increases cell survival (Figure 2). Identifying these risky or protective lncRNAs and viability by activating NF-κB signaling, which accom- should help predict prognosis in glioma. Determining panies the acquisition of TMZ resistance by glioma cells the dysregulation statuses of lncRNAs associated with (Duan et al. 2018). TMZ resistance should aid the development of promis- In glioma, increased TMZ resistance, conferred by the ing pharmacological targets. Therefore, therapeutic upregulations of risky lncRNAs such as H19, MALAT1, strategies targeting the dysregulation of lncRNA accord- PVT1, and SBF2-AS1 (Figure 1), is likely to play a key ing to IDH mutation status will help improve the prog- role in the therapeutic efficacy of surgical resection noses of glioma patients. ANIMAL CELLS AND SYSTEMS 25 Chowdhury R, Yeoh KK, Tian Y-M, Hillringhaus L, Bagg EA, Rose Disclosure statement NR, Leung IKH, Li XS, Woon ECY, Yang M, et al. 2011. The No potential conflict of interest was reported by the author(s). oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases. EMBO Rep. 12(5):463–469. doi:10.1038/ embor.2011.43. Chu C, Zhang QC, da Rocha ST, Flynn RA, Bharadwaj M, References Calabrese JM, Magnuson T, Heard E, Chang HY. 2015. Systematic discovery of xist RNA binding proteins. Cell. Abdel-Wahab N, Wicks SJ, Mason RM, Chantry A. 2002. Decorin 161(2):404–416. doi:10.1016/j.cell.2015.03.025. suppresses transforming growth factor-beta-induced Chu E, Callender MA, Farrell MP, Schmitz JC. 2003. Thymidylate expression of plasminogen activator inhibitor-1 in human synthase inhibitors as anticancer agents: from bench to mesangial cells through a mechanism that involves Ca2 bedside. Cancer Chemother Pharmacol. 52(Suppl 1):S80– +-dependent phosphorylation of Smad2 at serine-240. S89. doi:10.1007/s00280-003-0625-9. Biochem J. 362(Pt 3):643–649. doi:10.1042/0264- Dang L, White DW, Gross S, Bennett BD, Bittinger MA, 6021:3620643. Driggers EM, Fantin VR, Jang HG, Jin S, Keenan MC, Bazzoli E, Pulvirenti T, Oberstadt MC, Perna F, Wee B, Schultz N, et al. 2009. Cancer-associated IDH1 mutations produce 2- Huse JT, Fomchenko EI, Voza F, Tabar V, et al. 2012. MEF pro- hydroxyglutarate. Nature. 462(7274):739–744. doi:10.1038/ motes stemness in the pathogenesis of gliomas. Cell Stem nature08617. Cell. 11(6):836–844. doi:10.1016/j.stem.2012.09.012. de Cremoux P, Jourdan-Da-Silva N, Couturier J, Tran-Perennou Benjamin EJ, Rice KM, Arking DE, Pfeufer A, van Noord C, Smith C, Schleiermacher G, Fehlbaum P, Doz F, Mosseri V, Delattre AV, Schnabel RB, Bis JC, Boerwinkle E, Sinner MF, et al. 2009. O, Klijanienko J, et al. 2007. Role of chemotherapy resistance Variants in ZFHX3 are associated with atrial fibrillation in genes in outcome of neuroblastoma. Pediatr Blood Cancer. individuals of European ancestry. Nat Genet. 41(8):879– 48(3):311–317. doi:10.1002/pbc.20853. 881. doi:10.1038/ng.416. Do H, Kim D, Kang J, Son B, Seo D, Youn H, Youn B, Kim W. 2019. Cancer Genome Atlas Research Network, Brat DJ, Verhaak TFAP2C increases cell proliferation by downregulating RGW, Aldape KD, Yung WKA, Salama SR, Cooper LAD, GADD45B and PMAIP1 in non-small cell lung cancer cells. Rheinbay E, Miller CR, Vitucci M, et al. 2015. Biol Res. 52(1):35. doi:10.1186/s40659-019-0244-5. Comprehensive, integrative genomic analysis of diffuse Do H, Kim W. 2018. Roles of oncogenic long non-coding RNAs lower-grade gliomas. N Engl J Med. 372(26):2481–2498. in cancer development. Genomics Inform. 16(4):e18. doi:10. doi:10.1056/NEJMoa1402121. 5808/GI.2018.16.4.e18. Cai T, Liu Y, Xiao J. 2018. Long noncoding RNA MALAT1 knock- Dong N, Guo J, Han S, Bao L, Diao Y, Lin Z. 2019. Positive feed- down reverses chemoresistance to temozolomide via pro- back loop of lncRNA HOXC-AS2/miR-876-5p/ZEB1 to regu- moting microRNA-101 in glioblastoma. Cancer Med. 7 late EMT in glioma. Onco Targets Ther. 12:7601–7609. (4):1404–1415. doi:10.1002/cam4.1384. doi:10.2147/OTT.S216134. Chae Y, Roh J, Kim W. 2021. The roles played by long non- Duan S, Li M, Wang Z, Wang L, Liu Y. 2018. H19 induced by oxi- coding RNAs in glioma resistance. Int J Mol Sci. 22(13) dative stress confers temozolomide resistance in human doi:10.3390/ijms22136834. glioma cells via activating NF-κB signaling. Onco Targets Chang Y-Z, Chai R-C, Pang B, Chang X, SY A, Zhang K-N, Jiang T, Ther. 11:6395–6404. doi:10.2147/OTT.S173244. Wang Y-Z. 2021. METTL3 enhances the stability of MALAT1 Gong R, Li Z-Q, Fu K, Ma C, Wang W, Chen J-C. 2021. Long non- with the assistance of HuR via m6A modification and acti- coding RNA PVT1 promotes stemness and temozolomide vates NF-κB to promote the malignant progression of IDH- resistance through miR-365/ELF4/SOX2 axis in glioma. Exp wildtype glioma. Cancer Lett. 511:36–46. doi:10.1016/j. Neurobiol. 30(3):244–255. doi:10.5607/en20060. canlet.2021.04.020. González-Gómez P, Anselmo NP, Mira H. 2014. BMPs as thera- Chen J, Han P, Dahiya S. 2022a. Glioblastoma: changing con- peutic targets and biomarkers in astrocytic glioma. Biomed cepts in the WHO CNS5 classification. Indian J Pathol Res Int. 2014:549742. doi:10.1155/2014/549742. Microbiol. 65(Supplement):S24–S32. doi:10.4103/ijpm. Gudbjartsson DF, Holm H, Gretarsdottir S, Thorleifsson G, ijpm_1109_21. Walters GB, Thorgeirsson G, Gulcher J, Mathiesen EB, Chen K, Wang X, Wei B, Sun R, Wu C, Yang H. 2022b. LncRNA Njølstad I, Nyrnes A, et al. 2009. A sequence variant in SNHG6 promotes glycolysis reprogramming in hepatocellu- ZFHX3 on 16q22 associates with atrial fibrillation and lar carcinoma by stabilizing the BOP1 protein. Anim Cells ischemic stroke. Nat Genet. 41(8):876–878. doi:10.1038/ng. Syst (Seoul). 26(6):369–379. doi:10.1080/19768354.2022. Gudjonsson T, Altmeyer M, Savic V, Toledo L, Dinant C, Grøfte Chen W, Xu X-K, Li J-L, Kong K-K, Li H, Chen C, He J, Wang F, Li P, M, Bartkova J, Poulsen M, Oka Y, Bekker-Jensen S, et al. 2012. Ge X-S, Li F-C. 2017. MALAT1 is a prognostic factor in glio- TRIP12 and UBR5 suppress spreading of chromatin ubiquity- blastoma multiforme and induces chemoresistance to temo- lation at damaged chromosomes. Cell. 150(4):697–709. zolomide through suppressing miR-203 and promoting doi:10.1016/j.cell.2012.06.039. thymidylate synthase expression. Oncotarget. 8(14):22783– Hausinger RP. 2004. FeII/alpha-ketoglutarate-dependent 22799. doi:10.18632/oncotarget.15199. hydroxylases and related enzymes. Crit Rev Biochem Mol Chen Y, Guo Y, Chen H, Ma F. 2020. Long non-coding RNA Biol. 39(1):21–68. doi:10.1080/10409230490440541. expression profiling identifies a four-long non-coding RNA Hayward D, Cole PA. 2016. LSD1 Histone Demethylase Assays prognostic signature for isocitrate dehydrogenase mutant and Inhibition; Methods Enzymol. 573:261–278. doi:10. glioma. Front Neurol. 11:573264. doi:10.3389/fneur.2020. 1016/bs.mie.2016.01.020. 573264. 26 J. ROH ET AL. He Z, Long J, Yang C, Gong B, Cheng M, Wang Q, Tang J. 2020. Lee SY. 2016. Temozolomide resistance in glioblastoma multi- LncRNA DGCR5 plays a tumor-suppressive role in glioma via forme. Genes Dis. 3(3):198–210. doi:10.1016/j.gendis.2016. the miR-21/Smad7 and miR-23a/PTEN axes. Aging. 12 04.007. (20):20285–20307. doi:10.18632/aging.103800. Leeper HE, Caron AA, Decker PA, Jenkins RB, Lachance DH, Horbinski C, Berger T, Packer RJ, Wen PY. 2022. Clinical impli- Giannini C. 2015. IDH mutation, 1p19q codeletion and cations of the 2021 edition of the WHO classification of ATRX loss in WHO grade II gliomas. Oncotarget. 6 central nervous system tumours. Nat Rev Neurol. 18 (30):30295–30305. doi:10.18632/oncotarget.4497. (9):515–529. doi:10.1038/s41582-022-00679-w. Leighton F, Poole B, Lazarow PB, de Duve C. 1969. The synthesis Huang G-H, Pei Y-C, Yang L, Mou K-J, Tang J-H, Xiang Y, Liu J, Lv and turnover of rat liver peroxisomes. I. fractionation of per- S-Q. 2020. Integrative transcriptome analysis identified a oxisome proteins. J Cell Biol. 41(2):521–535. doi:10.1083/jcb. BMP signaling pathway-regulated lncRNA AC068643.1 in 41.2.521. IDH mutant and wild-type glioblastomas. Oncol Lett. 20 Li H, Yuan X, Yan D, Li D, Guan F, Dong Y, Wang H, Liu X, Yang B. (1):75–84. doi:10.3892/ol.2020.11542. 2017. Long non-coding RNA MALAT1 decreases the sensi- Hurley JH, Dean AM, Koshland DE, Stroud RM. 1991. Catalytic tivity of resistant glioblastoma cell lines to temozolomide. mechanism of NADP(+)-dependent isocitrate dehydrogen- Cell Physiol Biochem. 42(3):1192–1201. doi:10.1159/ ase: implications from the structures of magnesium-isoci- 000478917. trate and NADP+ complexes. Biochemistry. 30(35):8671– Li J, Chen Y, Zhao J, Kong F, Zhang Y. 2011. miR-203 reverses 8678. doi:10.1021/bi00099a026. chemoresistance in p53-mutated colon cancer cells Itsumi M, Inoue S, Elia AJ, Murakami K, Sasaki M, Lind EF, Brenner through downregulation of Akt2 expression. Cancer Lett. D, Harris IS, Chio IIC, Afzal S, et al. 2015. Idh1 protects murine 304(1):52–59. doi:10.1016/j.canlet.2011.02.003. hepatocytes from endotoxin-induced oxidative stress by Li R, Chen W, Mao P, Wang J, Jing J, Sun Q, Wang M, Yu X. 2021a. regulating the intracellular NADP(+)/NADPH ratio. Cell Identification of a three-long non-coding RNA signature for Death Differ. 22(11):1837–1845. doi:10.1038/cdd.2015.38. predicting survival of temozolomide-treated isocitrate dehy- Jiang P, Wang P, Sun X, Yuan Z, Zhan R, Ma X, Li W. 2016. drogenase mutant low-grade gliomas. Exp Biol Med Knockdown of long noncoding RNA H19 sensitizes human (Maywood). 246(2):187–196. doi:10.1177/1535370220962715. glioma cells to temozolomide therapy. Onco Targets Ther. Li Z, Meng X, Wu P, Zha C, Han B, Li L, Sun N, Qi T, Qin J, 9:3501–3509. doi:10.2147/OTT.S96278. Zhang Y, et al. 2021b. Glioblastoma cell-derived lncRNA- Jiang W, Wang S, Xiao M, Lin Y, Zhou L, Lei Q, Xiong Y, Guan K- containing exosomes induce microglia to produce comp- L, Zhao S. 2011. Acetylation regulates gluconeogenesis by lement C5, promoting chemotherapy resistance. Cancer promoting PEPCK1 degradation via recruiting the UBR5 ubi- Immunol Res. 9(12):1383–1399. doi:10.1158/2326-6066.CIR- quitin ligase. Mol Cell. 43(1):33–44. doi:10.1016/j.molcel. 21-0258. 2011.04.028. Lin J, Lin Y, Fan L, Kuang W, Zheng L, Wu J, Shang P, Wang Q, Karreth FA, Tay Y, Perna D, Ala U, Tan SM, Rust AG, DeNicola G, Tan J. 2016. miR-203 inhibits cell proliferation and promotes Webster KA, Weiss D, Perez-Mancera PA, et al. 2011. In vivo cisplatin induced cell death in tongue squamous cancer. identification of tumor- suppressive PTEN ceRNAs in an Biochem Biophys Res Commun. 473(2):382–387. doi:10. oncogenic BRAF-induced mouse model of melanoma. Cell. 1016/j.bbrc.2016.02.105. 147(2):382–395. doi:10.1016/j.cell.2011.09.032. Liu ZZ, Tian YF, Wu H, Ouyang SY, Kuang WL. 2020. LncRNA Katoh M. 2016. Mutation spectra of histone methyltransferases H19 promotes glioma angiogenesis through miR-138/HIF- with canonical SET domains and EZH2-targeted therapy. 1α/VEGF axis. Neoplasma. 67(1):111–118. doi:10.4149/neo_ Epigenomics. 8(2):285–305. doi:10.2217/epi.15.89. 2019_190121N61. Koh H-J, Lee S-M, Son B-G, Lee S-H, Ryoo ZY, Chang K-T, Park J- Loenarz C, Schofield CJ. 2008. Expanding chemical biology of W, Park D-C, Song BJ, Veech RL, et al. 2004. Cytosolic NADP 2-oxoglutarate oxygenases. Nat Chem Biol. 4(3):152–156. +-dependent isocitrate dehydrogenase plays a key role in doi:10.1038/nchembio0308-152. lipid metabolism. J Biol Chem. 279(38):39968–39974. Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella- doi:10.1074/jbc.M402260200. Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues Komori T. 2022. Grading of adult diffuse gliomas according to P, Ellison DW. 2016. The 2016 world health organization the 2021 WHO classification of tumors of the central nervous classification of tumors of the central nervous system: a system. Lab Invest. 102(2):126–133. doi:10.1038/s41374-021- summary. Acta Neuropathol. 131(6):803–820. doi:10.1007/ 00667-6. s00401-016-1545-1. Kuang Y, Bing Z, Jin X, Li Q. 2021. LncRNA H19 upregulation Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella- participates in the response of glioma cells to radiation. Branger D, Hawkins C, Ng HK, Pfister SM, Reifenberger G, Biomed Res Int. 2021:1728352. doi:10.1155/2021/1728352. et al. 2021. The 2021 WHO classification of tumors of the Lee S, Kim YN, Im D, Cho SH, Kim J, Kim J-H, Kim K. 2021. DNA central nervous system: a summary. Neuro Oncol. 23 methylation and gene expression patterns are widely (8):1231–1251. doi:10.1093/neuonc/noab106. altered in fetal growth restriction and associated with FGR Lu C, Ward PS, Kapoor GS, Rohle D, Turcan S, Abdel-Wahab O, development. Anim Cells Syst (Seoul). 25(3):128–135. Edwards CR, Khanin R, Figueroa ME, Melnick A, et al. 2012. doi:10.1080/19768354.2021.1925741. IDH mutation impairs histone demethylation and results in Lee SH, Jo SH, Lee SM, Koh HJ, Song H, Park JW, Lee WH, Huh a block to cell differentiation. Nature. 483(7390):474–478. TL. 2004. Role of NADP+-dependent isocitrate dehydrogen- doi:10.1038/nature10860. ase (NADP+-ICDH) on cellular defence against oxidative Lu C, Wei Y, Wang X, Zhang Z, Yin J, Li W, Chen L, Lyu X, Shi Z, injury by gamma-rays. Int J Radiat Biol. 80(9):635–642. Yan W, You Y. 2020. DNA-methylation-mediated activating doi:10.1080/09553000400007680. of lncRNA SNHG12 promotes temozolomide resistance in ANIMAL CELLS AND SYSTEMS 27 glioblastoma. Mol Cancer. 19(1):28. doi:10.1186/s12943-020- Shao Y, Ye M, Li Q, Sun W, Ye G, Zhang X, Yang Y, Xiao B, Guo J. 1137-5. 2016. LncRNA-RMRP promotes carcinogenesis by acting as a Ludwig K, Kornblum HI. 2017. Molecular markers in glioma. J miR-206 sponge and is used as a novel biomarker for gastric Neurooncol. 134(3):505–512. doi:10.1007/s11060-017-2379-y. cancer. Oncotarget. 7(25):37812–37824. doi:10.18632/ MacDonald MJ, Brown LJ, Longacre MJ, Stoker SW, Kendrick oncotarget.9336. MA, Hasan NM. 2013. Knockdown of both mitochondrial iso- Sun H, Yin L, Li S, Han S, Song G, Liu N, Yan C. 2013. Prognostic citrate dehydrogenase enzymes in pancreatic beta cells inhi- significance of IDH mutation in adult low-grade gliomas: a bits insulin secretion. Biochim Biophys Acta. 1830(11):5104– meta-analysis. J Neurooncol. 113(2):277–284. doi:10.1007/ 5111. doi:10.1016/j.bbagen.2013.07.013. s11060-013-1107-5. Marín-Béjar O, Marchese FP, Athie A, Sánchez Y, González J, Tay Y, Rinn J, Pandolfi PP. 2014. The multilayered complexity of Segura V, Huang L, Moreno I, Navarro A, Monzó M, et al. ceRNA crosstalk and competition. Nature. 505(7483):344– 2013. Pint lincRNA connects the p53 pathway with epige- 352. doi:10.1038/nature12986. netic silencing by the polycomb repressive complex 2. Tian T, Mingyi M, Qiu X, Qiu Y. 2016. MicroRNA-101 reverses Genome Biol. 14(9):R104. doi:10.1186/gb-2013-14-9-r104. temozolomide resistance by inhibition of GSK3β in glioblas- McNamara MG, Sahebjam S, Mason WP. 2013. Emerging bio- toma. Oncotarget. 7(48):79584–79595. doi:10.18632/ markers in glioblastoma. Cancers (Basel). 5(3):1103–1119. oncotarget.12861. doi:10.3390/cancers5031103. Turcan S, Rohle D, Goenka A, Walsh LA, Fang F, Yilmaz E, Mercer TR, Dinger ME, Mattick JS. 2009. Long non-coding RNAs: Campos C, Fabius AWM, Lu C, Ward PS, et al. 2012. IDH1 insights into functions. Nat Rev Genet. 10(3):155–159. doi:10. mutation is sufficient to establish the glioma hypermethyla- 1038/nrg2521. tor phenotype. Nature. 483(7390):479–483. doi:10.1038/ Nobusawa S, Watanabe T, Kleihues P, Ohgaki H. 2009. IDH1 nature10866. mutations as molecular signature and predictive factor of Waitkus MS, Diplas BH, Yan H. 2018. Biological role and thera- secondary glioblastomas. Clin Cancer Res. 15(19):6002– peutic potential of IDH mutations in cancer. Cancer Cell. 34 6007. doi:10.1158/1078-0432.CCR-09-0715. (2):186–195. doi:10.1016/j.ccell.2018.04.011. Peng Z, Liu C, Wu M. 2018. New insights into long noncoding Ward PS, Patel J, Wise DR, Abdel-Wahab O, Bennett BD, Coller RNAs and their roles in glioma. Mol Cancer. 17(1):61. doi:10. HA, Cross JR, Fantin VR, Hedvat CV, Perl AE, et al. 2010. The 1186/s12943-018-0812-2. common feature of leukemia-associated IDH1 and IDH2 Pyko IV, Nakada M, Sabit H, Teng L, Furuyama N, Hayashi Y, mutations is a neomorphic enzyme activity converting Kawakami K, Minamoto T, Fedulau AS, Hamada J. 2013. alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell. 17 Glycogen synthase kinase 3β inhibition sensitizes human (3):225–234. doi:10.1016/j.ccr.2010.01.020. glioblastoma cells to temozolomide by affecting O6-methyl- Weller M, Wick W, Aldape K, Brada M, Berger M, Pfister SM, guanine DNA methyltransferase promoter methylation via Nishikawa R, Rosenthal M, Wen PY, Stupp R, Reifenberger c-Myc signaling. Carcinogenesis. 34(10):2206–2217. doi:10. G. 2015. Glioma. Nat Rev Dis Primers. 1:15017. doi:10.1038/ 1093/carcin/bgt182. nrdp.2015.17. Rahman Z, Bazaz Mohd R, Devabattula G, Khan Mohd A, Wick W, Hartmann C, Engel C, Stoffels M, Felsberg J, Godugu C. 2021. Targeting H3K9 methyltransferase G9a Stockhammer F, Sabel MC, Koeppen S, Ketter R, and its related molecule GLP as a potential therapeutic strat- Meyermann R, et al. 2009. NOA-04 randomized phase III egy for cancer. J Biochem Mol Toxicol. 35(3). doi:10.1002/jbt. trial of sequential radiochemotherapy of anaplastic glioma 22674. with procarbazine, lomustine, and vincristine or temozolo- Reitman ZJ, Yan H. 2010. Isocitrate dehydrogenase 1 and 2 mide. J Clin Oncol. 27(35):5874–5880. doi:10.1200/JCO. mutations in cancer: alterations at a crossroads of cellular 2009.23.6497. metabolism. J Natl Cancer Inst. 102(13):932–941. doi:10. Wu F, Zhao Z, Chai R, Liu Y, Wang K, Wang Z, Li G, Huang R, 1093/jnci/djq187. Jiang H, Zhang K. 2018. Expression profile analysis of anti- Rickert DE. 1987. Metabolism of nitroaromatic compounds. Drug sense long non-coding RNA identifies WDFY3-AS2 as a prog- Metab Rev. 18(1):23–53. doi:10.3109/03602538708998299. nostic biomarker in diffuse glioma. Cancer Cell Int. 18:107. Sachdeva R, Wu M, Johnson K, Kim H, Celebre A, Shahzad U, doi:10.1186/s12935-018-0603-2. Graham MS, Kessler JA, Chuang JH, Karamchandani J, et al. Xiong Z, Wang L, Wang Q, Yuan Y. 2018. LncRNA MALAT1/ 2019. BMP signaling mediates glioma stem cell quiescence miR-129 axis promotes glioma tumorigenesis by targeting and confers treatment resistance in glioblastoma. Sci Rep. SOX2. J Cell Mol Med. 22(8):3929–3940. doi:10.1111/jcmm. 9(1):14569. doi:10.1038/s41598-019-51270-1. 13667. Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP. 2011. A ceRNA Xu W, Yang H, Liu Y, Yang Y, Ping W, Kim S-H, Ito S, Yang C, Pu hypothesis: the rosetta stone of a hidden RNA language? W, Xiao M-T, et al. 2011. Oncometabolite 2-hydroxyglutarate Cell. 146(3):353–358. doi:10.1016/j.cell.2011.07.014. is a competitive inhibitor of α-ketoglutarate-dependent Seo D, Kim D, Chae Y, Kim W. 2020. The ceRNA network of dioxygenases. Cancer Cell. 19(1):17–30. doi:10.1016/j.ccr. lncRNA and miRNA in lung cancer. Genomics Inform. 18(4): 2010.12.014. e36. doi:10.5808/GI.2020.18.4.e36. Xu X, Zhao J, Xu Z, Peng B, Huang Q, Arnold E, Ding J. 2004. Seo D, Kim D, Kim W. 2019. Long non-coding RNA linc00152 Structures of human cytosolic NADP-dependent isocitrate acting as a promising oncogene in cancer progression. dehydrogenase reveal a novel self-regulatory mechanism Genomics Inform. 17(4):e36. doi:10.5808/GI.2019.17.4.e36. of activity. J Biol Chem. 279(32):33946–33957. doi:10.1074/ Seo D, Roh J, Chae Y, Kim W. 2021. Gene expression profiling jbc.M404298200. after LINC00472 overexpression in an NSCLC cell line1. Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan W, Cancer Biomark. 32(2):175–188. doi:10.3233/CBM-210242. Kos I, Batinic-Haberle I, Jones S, Riggins GJ, et al. 2009. 28 J. ROH ET AL. IDH1 and IDH2 mutations in gliomas. N Engl J Med. 360 Zhang Y, Zhou S, Yan H, Xu D, Chen H, Xiao-yan W, Xiao W, Liu (8):765–773. doi:10.1056/NEJMoa0808710. Y, Zhang L, Wang S, et al. 2016. miR-203 inhibits proliferation Yang B, Zhong C, Peng Y, Lai Z, Ding J. 2010. Molecular mech- and self-renewal of leukemia stem cells by targeting survivin anisms of “off-on switch” of activities of human IDH1 by and Bmi-1. Sci Rep. 6:19995. doi:10.1038/srep19995. tumor-associated mutation R132H. Cell Res. 20(11):1188– Zhang Z, Yin J, Lu C, Wei Y, Zeng A, You Y. 2019b. Exosomal 1200. doi:10.1038/cr.2010.145. transfer of long non-coding RNA SBF2-AS1 enhances che- Youssef G, Miller JJ. 2020. Lower grade gliomas. Curr moresistance to temozolomide in glioblastoma. J Exp Clin Neurol Neurosci Rep. 20(7):21. doi:10.1007/s11910-020- Cancer Res. 38(1):166. doi:10.1186/s13046-019-1139-6. 01040-8. Zhao S, Lin Y, Xu W, Jiang W, Zha Z, Wang P, Yu W, Li Z, Gong L, Yu H, Zheng J, Liu X, Xue Y, Shen S, Zhao L, Li Z, Liu Y. 2017. Peng Y, et al. 2009. Glioma-derived mutations in IDH1 dom- Transcription factor NFAT5 promotes glioblastoma cell- inantly inhibit IDH1 catalytic activity and induce HIF-1alpha. driven angiogenesis via SBF2-AS1/miR-338-3p-mediated Science. 324(5924):261–265. doi:10.1126/science.1170944. EGFL7 expression change. Front Mol Neurosci. 10:301. Zhou Q, Liu Z-Z, Wu H, Kuang W-L. 2022. LncRNA H19 pro- doi:10.3389/fnmol.2017.00301. motes cell proliferation, migration, and angiogenesis of Zhang J, Zhang P, Wang L, Piao H, Ma L. 2014. Long non-coding glioma by regulating Wnt5a/β-catenin pathway via target- RNA HOTAIR in carcinogenesis and metastasis. Acta Biochim ing miR-342. Cell Mol Neurobiol. 42(4):1065–1077. doi:10. Biophys Sin (Shanghai). 46(1):1–5. doi:10.1093/abbs/ 1007/s10571-020-00995-z. gmt117. Zhou Y, Hu X-W, Yang S-J, Yu Z. 2020. Knockdown of Zhang Q, Liu X-J, Li Y, Ying X-W, Chen L. 2021. Prognostic value lncRNAZFAS1 suppresses cell proliferation and metastasis of immune-related lncRNA SBF2-AS1 in diffuse lower-grade in non-small cell lung cancer. Anim Cells Syst (Seoul). 24 glioma. Technol Cancer Res Treat. 20:15330338211011966. (2):107–113. doi:10.1080/19768354.2020.1736623. doi:10.1177/15330338211011966. Zou H, Wu L-X, Yang Y, Li S, Mei Y, Liu Y-B, Zhang L, Cheng Y, Zhang Y, Yang G, Luo Y. 2019a. Long non-coding RNA PVT1 Zhou H-H. 2017. lncRNAs PVT1 and HAR1A are prognosis promotes glioma cell proliferation and invasion by targeting biomarkers and indicate therapy outcome for diffuse miR-200a. Exp Ther Med. 17(2):1337–1345. doi:10.3892/etm. glioma patients. Oncotarget. 8(45):78767–78780. doi:10. 2018.7083. 18632/oncotarget.20226.

Journal

Animal Cells and SystemsTaylor & Francis

Published: Dec 11, 2023

Keywords: Isocitrate dehydrogenase; long non-coding RNA; glioma; temozolomide resistance

There are no references for this article.