Next Article in Journal
Small RNAs beyond Model Organisms: Have We Only Scratched the Surface?
Next Article in Special Issue
Multi-Cohort Transcriptomic Subtyping of B-Cell Acute Lymphoblastic Leukemia
Previous Article in Journal
Leaf Mutant 7 Encoding Heat Shock Protein OsHSP40 Regulates Leaf Size in Rice
Previous Article in Special Issue
Lineage Conversion in Pediatric B-Cell Precursor Acute Leukemia under Blinatumomab Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Linear and Circular Long Non-Coding RNAs in Acute Lymphoblastic Leukemia: From Pathogenesis to Classification and Treatment

1
The Affiliated Sir Run Run Shaw Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
2
Cancer Center, Zhejiang University, Hangzhou 310058, China
3
NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
4
Department of Hematology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
5
Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(8), 4442; https://doi.org/10.3390/ijms23084442
Submission received: 28 March 2022 / Revised: 13 April 2022 / Accepted: 15 April 2022 / Published: 18 April 2022
(This article belongs to the Special Issue Molecular Research on Acute Lymphoblastic Leukemia 2.0)

Abstract

:
The coding regions account for only a small part of the human genome, and the remaining vast majority of the regions generate large amounts of non-coding RNAs. Although non-coding RNAs do not code for any protein, they are suggested to work as either tumor suppressers or oncogenes through modulating the expression of genes and functions of proteins at transcriptional, posttranscriptional and post-translational levels. Acute Lymphoblastic Leukemia (ALL) originates from malignant transformed B/T-precursor-stage lymphoid progenitors in the bone marrow (BM). The pathogenesis of ALL is closely associated with aberrant genetic alterations that block lymphoid differentiation and drive abnormal cell proliferation as well as survival. While treatment of pediatric ALL represents a major success story in chemotherapy-based elimination of a malignancy, adult ALL remains a devastating disease with relatively poor prognosis. Thus, novel aspects in the pathogenesis and progression of ALL, especially in the adult population, need to be further explored. Accumulating evidence indicated that genetic changes alone are rarely sufficient for development of ALL. Recent advances in cytogenic and sequencing technologies revealed epigenetic alterations including that of non-coding RNAs as cooperating events in ALL etiology and progression. While the role of micro RNAs in ALL has been extensively reviewed, less attention, relatively, has been paid to other non-coding RNAs. Herein, we review the involvement of linear and circular long non-coding RNAs in the etiology, maintenance, and progression of ALL, highlighting the contribution of these non-coding RNAs in ALL classification and diagnosis, risk stratification as well as treatment.

1. Introduction

Ribonucleic acid (RNA) is well-known as functioning in gene transcription and translation to generate numerous proteins, which exert various biological functions. While the coding regions only account for around 1.5% of the entire human genome, the remaining non-coding regions are also extensively transcribed to produce large amount of non-coding RNAs (ncRNAs) [1,2]. The pre-mRNA splicing, processing as well as other cellular events also generate some non-coding RNAs such as circular RNAs (circRNAs) [3]. According to reports from the Encyclopedia of DNA Elements (ENCODE) project, up to 80% of the human genome has the capacity to transcribe into ncRNAs [4,5]. Based on their size and/or location and/or interacting partners, ncRNAs are classified into several types including transfer RNAs (tRNAs), ribosome RNAs (rRNAs), small nucleolar RNAs (snoRNAs), small nuclear RNAs (snRNAs), microRNAs (miRNAs), small interfering RNAs (siRNAs), long non-coding RNAs (lncRNAs), circular RNA (circRNAs), piwi-interacting RNAs (piRNAs), and enhancer RNAs (eRNAs) (Figure 1) [2,6]. Accumulating evidence indicated that different types of ncRNAs are important players in modulation of complex molecular and cellular processes [2,6,7]. Indeed, although there is a lack of open reading frames (ORFs) to code for proteins, these ncRNAs exert important functions in the transcription of genes, as well as RNA splicing, chemical modification, stability, and translation among others (Figure 1). As emerging sculptures of the epigenetic landscape, gaining comprehensive knowledge on the features and functional versatilities of these molecules in normal and cancerous conditions might help in directing new therapeutic interventions in the future.
Acute lymphoblastic leukemia (ALL) is characterized by differentiation-block at B/T-precursor stage lymphoid progenitors that can invade BM, peripheral blood, and extramedullary sites (e.g., the central nervous system) [8]. These committed cells acquire resistance to apoptosis and differentiation, and display hallmark cancer phenotypes including continuous self-renewal, abnormal proliferation, and metastasis among others. Diagnosis is established by the presence of more than 20% lymphoblasts in the BM or peripheral blood [9,10]. According to the committed cells of origin, ALL is classified into B-cell ALL (B-ALL), and T-cell ALL (T-ALL). B-ALL constitutes approximately 80–85% of total ALL cases with T-ALL comprising the rest [10]. B-ALL is further divided into many genetic subtypes according to the involved chromosomal/genetic abnormalities such as chromatin number alterations (e.g., high hyperdiploid, near haploid, low hypodiploid), rearrangements (e.g., MLL, ZNF384, MEF2D, DUX4), deletions (e.g., ERG), fusion oncogene producing translocations (e.g., ETV6-RUNX1+, BCR-ABL1+, E2A-BPX1+, E2A-HLF+) and so on [8,11,12]. Meanwhile, T-ALL is classified into four main subtypes according to the characteristic oncogenic aberrations, namely TLX (TLX3 rearrangements or HOXA activating events), TLX1/NKX2.1 (LTX1 or NKX2.1 rearrangements), TAL/LMO (high expression of TAL1, TAL2, LMO1/2/3), and early thymocyte progenitor (ETP) /immature-ALL (high expression of LMO2, LYL1, HHEX, and BCL2) [10,13]. Such complexity of molecular and cytogenetic hallmarks determined heterogeneity of ALL, which is also reflected by the differing response of varying ALL subtypes to similar therapeutic regimens [8,14,15].
It is noteworthy that the incidence of ALL is highest among children aged 1–4 years and declines with aging with a slight increase after the age of 50 [8,16]. Thus, ALL represents the most frequent acute leukemia in the pediatric population, it is also the second frequent acute leukemia of adults [8,9,10]. The prognosis of ALL differs a lot among the pediatric and adult populations. With currently used state-of-the-art treatments, long-term remission of adult patients with ALL barely reaches 30–40% as compared to that of more than 80% in the pediatric ALL population [9,17]. Furthermore, relapse of ALL remains a leading cause of childhood cancer-related death [18]. Therefore, a comprehensive mechanistic understanding of the etiology and progression of ALL is of the utmost importance to develop better clinical options for management of refractory/relapsed ALL patients.
Recent developments in high-throughput sequencing technologies and bioinformatics tools have enabled comprehensive molecular characterization of various non-coding RNA expression profiles in normal and cancerous cells. Several types of ncRNAs including rRNAs, tRNAs, snRNAs, and snoRNAs are constitutively expressed in most cell types and essential for cell viability; thus, they are categorized as housekeeping ncRNAs [6,7]. Meanwhile, apart from the ncRNAs mentioned above, other types of ncRNAs such as miRNAs, lncRNAs, circRNAs, and piRNAs are classified as regulatory ncRNAs, which show tissue-specific as well as spatiotemporal expression patterns and function as tumor suppressors or oncogenes through modulating gene expression via interactions with other biomolecules (e.g., coding and non-coding RNAs, DNAs, and proteins) [6,7]. It is reported that ETV6-RUNX1 is present in B-cell progenitors years before the onset of ALL, suggesting genetic changes alone are not sufficient to initiate ALL [19,20]. A growing body of evidence indicated the involvement of regulatory ncRNAs including miRNAs, lncRNAs, and circRNAs as cooperating events of genetic alterations in ALL [21,22], among these, the role of miRNAs in ALL pathogenesis and treatment has been extensively reviewed elsewhere [21,22,23]. In this review, we mainly recapitulate the involvement of linear and circular long non-coding RNAs in the etiology, maintenance, and progression of ALL, providing a comprehensive picture to reveal the implications of these long non-coding RNAs in ALL diagnosis, risk stratification as well as treatment.

2. Current Challenges in ALL Treatment

ALL is a highly heterogeneous disease. In clinic, ALL cases are stratified into favorable-, intermediate-, and adverse-risk groups based on the cytogenetic profiles [9]. Frontline treatment of ALL conventionally includes sequential phases of induction, consolidation, intensification and maintenance chemotherapy with regimens containing vincristine, cyclophosphamide, anthracyclines, L-asparaginase, and glucocorticoids among others, accompanied by central nervous system (CNS) prophylactic intrathecal chemotherapy with methotrexate alone, or methotrexate combined with cytarabine and hydrocortisone/dexamethasone, with or without CNS irradiation [8,9]. Tyrosine kinase inhibitors (TKIs, e.g., imatinib, dasatinib and ponatinib) are used in combination with the chemotherapy regimen for BCR-ABL1+ ALL (Ph+ ALL) patients [8,24]. For eligible patients (mostly MRD positive non-adolescent adult patients), allogenic stem cell transplantation (Allo-SCT) is recommended [8,25]. In addition, novel agents including monoclonal antibody drugs, proteasome inhibitors, JAK inhibitors, PI3K-mTOR inhibitors, and chimeric antigen receptor (CAR) T-cell therapy were also evaluated in clinical trials for ALL [8,26,27]. These novel agents have shown certain potential in improving ALL patient disease-free survival (DFS) when used in combination with conventional chemotherapy [8,24,25,26,27]. Nevertheless, obtaining long-term remission/cure in relapsed or refractory ALL patients is a current need.
Treatment of pediatric ALL represents a major success story and paradigm of malignant cancer elimination by dose-intensified chemotherapy regimens [28]. This is attributable to the improved risk stratification based on genetics as well as clinical characteristics at diagnosis, optimized chemotherapy regimens according to early treatment response as measured by minimal residual disease (MRD), and better tolerance of children to the chemotherapy regimens than adults [8,9,28,29]. However, relapsed ALL is still the second leading cause of childhood cancer-related death [18]. In addition, more than 50% of adult ALL patients who achieved a complete remission will ultimately relapse, and about 10% of adult ALL patients are initially refractory to the induction chemotherapy [30]. B-ALL subtypes associated with low hypodiploid (32–39 chromosomes), BCR-ABL+, and MLL (KMT2A) rearrangements, as well as T-ALL subtypes associated with HOXA activating events, mutations in IL-7R signaling molecules (IL7R, JAK1/3, STAT5B, N/KRAS, AKT), PTEN-inactivating events, and DNMT3A mutations among others develop refractory disease and show very poor prognosis [8,10,13,31]. There are few effective therapeutic approaches for relapsed/refractory ALL. One exception is the Ph+ ALL (BCR-ABL1+ ALL), which was once the most aggressive genetic abnormality in ALL and displayed very poor prognosis for both children and adults [32]. With the development and application of BCR-ABL1-targeted TKIs, the DFS of Ph+ ALL patients increased from around 20% to more than 60%, but there is still huge room for improvement [24,33]. Therefore, constant updates in therapeutic options are required to improve the survival rate of ALL patients.
CNS is the major site for the extramedullary invasion of most acute leukemias including ALL and the survival rate of CNS-infiltrated leukemia is rather poor [34,35]. In the early era of ALL treatment, CNS-targeted therapy was not included in the regimen, leading to as high as 80% of CNS relapse in all patients [36,37]. Later on, CNS prophylactic intrathecal chemotherapy with the ability to penetrate the blood–brain barrier was introduced into ALL treatment and largely reduced the overall risk of CNS relapse to 5% [38]. Notably, around 30–40% of relapsed ALL patients show CNS involvement, albeit many of them achieve a second complete remission (CR) [39]. The CNS-infiltrated ALL patients showing resistance to intrathecal chemotherapy respond well to the intracranial irradiation but face substantial neurotoxicity risks [40]. Common risk factors of CNS infiltration in ALL includes age, T-cell lineage (T-ALL), hyperleukocytosis, and some specific chromosomal aberrations such as MLL rearrangement, BCR-ABL1, and E2A-BPX1 fusion genes etc. [41]. In contrast to the role of qPCR-based MRD detection for monitoring BM responses and guiding risk stratification-based treatment adjustments [29,42], feasible and facile strategies for the detailed evaluation of CNS response are still lacking [43]. Thus, identification and comprehensive characterization of relapse (including CNS relapse) or refractory prone clones might offer precious opportunities to improve treatment outcomes in ALL patients.

3. Biogenesis and Biological Functions of lncRNAs

LncRNAs are characterized as ncRNA species of a size greater than 200 nucleotides (nt) without significant protein-coding capacity, which are yielded from eukaryotic RNA transcription and processing events [6,7]. This lenient definition determines lncRNAs as a large and highly heterogeneous family of transcripts with differing biogenesis pathways and genomic origins [44]. According to the estimates from ENCODE Project Consortium (GENCODE release 23), the human genome encodes more than 28,000 different lncRNAs [45]. Many lncRNAs display tissue-specific and spatiotemporal expression patterns, suggesting their important roles in cell fate determination. For instance, a previous study found that lncRNA X-inactive-specific transcript (Xist) knockout mice die early in embryogenesis as a result of two active X chromosomes induced extra-embryonic phenotype [46], highlighting the importance of lncRNAs in physio-pathological processes. Such distinct expression patterns and crucial functions of lncRNAs confers on them the potential of being used as feasible disease biomarkers as well as therapeutic targets.
Genomic origins of lncRNAs include promoter upstream regions, enhancers, intergenic/intragenic regions, and opposite strand of protein coding genes, among others [7,44]. LncRNAs are mostly transcribed by RNA polymerase II (Pol II), and less frequently by other RNA polymerases [44,47]. Thus, the resulting lncRNAs are often capped by 7-methyl guanosine (m7G) at their 5′ ends, polyadenylated at their 3′ ends, and even spliced similarly to mRNAs [44,48]. In addition, a considerable amount of lncRNAs are generated from long primary transcripts though other non-canonical RNA stabilizing pathways, such as RNase P cleavage to generate a mature 3′ end and capping by snoRNA-protein complexes at both ends or the 5′ end [49,50,51]. Taking advantage of flexible structure, lncRNAs interact with DNA, RNA, and proteins, thereby regulating gene expression and other bio-processes via affecting the chromatin state, transcription, biomolecular condensate formation, pre-mRNA splicing, mRNA stability, and translation (Figure 2) [44,45,48]. For instance, lncRNA ANRIL mediates polycomb repressive complex 1 (PRC1) and PRC2 recruitment to the promoters of its adjacent CDKN1B and CDKN2A genes, thereby controlling their expression and regulating cell senescence [52,53]. The lncRNA Xist inactivates a large proportion of the genes on the X chromosome via a complex interplay of protein interactors [54,55,56]. Recently, Statello et al. described various roles of lncRNAs in detail [2]. Together, these findings suggest that lncRNAs are feasible targets to intervene gene expression.
As compared to mRNAs, Pol II-transcribed lncRNAs are less efficiently processed, and a large proportion of such lncRNAs are retained in the nucleus [57,58]. These lncRNAs are either tethered to chromatin or participate in the formation of biomolecular condensates (BioMCs, also known as membranelles organelles) in the nucleus (Figure 2). For example, the long isoform of the lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) contributes to the organization and functions of paraspeckles via recruiting core proteins NONO and SFPQ to orchestrate the assembly of paraspeckles through liquid–liquid phase separation, regulating gene expression by sequestrating proteins or mRNAs with inverted repeats in their 3′ UTRs [59]. LncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) participates in the formation of nuclear speckles, regulating transcription and pre-mRNA splicing [60,61]. It is estimated that total RNA concentration in the nucleus is much higher than that in the cytoplasm [62], which is likely caused by, at least in part, the large amount of nuclear-retained lncRNAs. This characteristic high RNA concentration is reported to prevent aggregation of prion-like RNA binding proteins (RBPs) such as FUS and TDP43 in the nucleus [62,63], while FUS and TDP43 tend to aggregate in the cytoplasm probably due to less RNA concentration, causing neurodegenerative diseases including frontotemporal dementia and amyotrophic lateral sclerosis [62,64]. Interestingly, some lncRNAs with multiple exons are transported to the cytoplasm via nuclear RNA export factor 1 (NXF1) dependent pathway, interacting with RBPs and regulating mRNA stability as well as translation [48,65]. Accumulating evidence indicated that lncRNAs are widely associated with a majority of cancer types [66,67,68]. The aberrant expression of these lncRNA transcripts has been implicated in tumorigenesis, metastasis, cancer stage progression, and patient survival [68,69,70]. Importantly, these lncRNA molecules are easily targetable by antisense oligonucleotides (ASOs, suitable for downregulating nuclear lncRNAs) or siRNAs [48,71,72], suggesting that lncRNAs can be a class of druggable vulnerability in cancer.

4. LncRNAs in ALL Classification, Diagnosis, Pathogenesis, and Resistance to Treatment

Several studies have indicated that lncRNAs are associated with hematopoietic development [73,74,75,76], implying their potential involvement in hematological malignancies. Indeed, as important players in epigenetic regulation, lncRNAs have been demonstrated to function in various aspects of ALL (Table 1, Table 2 and Table 3) [77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116], which are summarized in this part.

4.1. LncRNAs in ALL Classification and Diagnosis

According to a study by Melo et al., the lncRNA expression profile showed significant alterations between different leukemia types and different subtypes of the same leukemia including ALL [117]. Another study found T-ALL subtype-specific lncRNA expression profiles, suggesting the involvement of lncRNAs as cooperators or downstream effectors during the corresponding oncogene-mediated leukemogenesis [118]. Alterations in chromatin and DNA modification also affect lncRNA expression profiles. It is reported that lncRNAs methylation per base pair shows a distinct profile between ALL and normal B-cell lymphoid precursors as well as among three sub clusters of ALL cases [119]. In human B-ALL samples, it was found that the lncRNA expression profile also correlates with cytogenetic abnormalities [102]. Likewise, many other studies also revealed subtype-specific and/or relapse-specific lncRNA signatures in ALL patent samples [77,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134]. These studies suggest that lncRNAs are associated with the pathogenesis of ALL and might be used as molecular markers for disease classification and diagnosis.

4.2. LncRNAs as Cooperating Events of Genetic Abnormalities in T-ALL Pathogenesis

T-ALL patients have a worse prognosis than those with B-ALL probably due to the less-known and more complex mechanism on its etiology, maintenance, and progression [8,27], suggesting the importance of exploring novel aspects in the pathogenesis and progression of T-ALL. Zhang et al. found that a lncRNA named T-ALL-related long non-coding RNA (T-ALL-R-LncR1) is markedly expressed in T-ALL Jurkat cells and 50% of T-ALL patient primary cells [79]. Importantly, knockdown of T-ALL-R-LncR1 led to apoptosis in Jurkat cells [79]. This finding suggests that the lncRNA T-ALL-R-LncR1 confers resistance to apoptosis, playing an oncogenic role in T-ALL development. It is well demonstrated that T cell acute lymphocytic leukemia protein 1 (TAL1, also known as SCL) is one of the most prevalent oncogenic transcription factors in T-ALL [10,13]. Generally, TAL1 coordinates with other transcription factors including GATA3, RUNX1, and MYB to regulate the expression of their downstream target genes in T-ALL cells [135]. Interestingly, TAL1 reportedly activated a subset of lncRNAs, some of which are regulated by GATA3,RUNX1, and MYB in a coordinated manner [133]; another subset of lncRNAs negatively regulated by TAL1 were also identified. Notably, the transcription factors T-cell leukemia homeobox 1 (TLX1) and TLX3 are key drivers of the TLX1/NKX2.1 and TLX subgroups of T-ALL, respectively [10,43]. A study showed that TLX1 directly regulates a set of lncRNAs, some of which are marked by super-enhancers [134], indicating the involvement of lncRNAs in TLX1-induced gene expression regulation. Intriguingly, some lncRNAs give rise to other ncRNA types following further processing. It was reported that TLX3 binds and transactivates lncRNA LINC00478, which is the host of miR125-b, to regulate miR-125b production, through which supports growth and invasiveness of T-ALL cells [81]. Collectively, these findings suggest that lncRNAs are crucial downstream effectors of major genetic abnormalities in the pathogenesis of T-ALL.
T-ALL is driven by oncogenic transcription factors (e.g., LTX1, LTX3 or HOXA, and TAL/LMO) that act along with secondary acquired mutations [10,13]. For instance, NOTCH1 activating mutations are identified in more than 50% of T-ALL cases, suggesting a key role in driving the disease [136]. A study by Durinck et al. established a novel lncRNA network that acts downstream of NOTCH1 during normal and malignant thymocyte development [137], specifically, they identified 40 lncRNAs that are positively regulated by NOTCH1 in both normal and malignant T lymphocytes, supporting an important role for these lncRNAs as downstream effectors in NOTCH1-regulated T-cell biology. Another study also uncovered a set of T-ALL-specific lncRNAs, many of which are directly regulated by the NOTCH1/RPBJκ activator complex [77]; the authors showed that one specific NOTCH-regulated lncRNA, LUNAR1, is required for efficient T-ALL growth in vitro and in vivo due to its ability to enhance insulin-like growth factor 1 (IGF1) receptor (IGF1R) mRNA level and sustain IGF1 signaling. Likewise, a lncRNA named NALT, which is located only 100 bp away from NOTCH1 gene, was upregulated in human NOTCH1-activated T-ALL samples [80]; increased expression of NALT dramatically promoted cell proliferation, while knockdown of NALT caused the opposite effect; mechanistically, nuclear located NALT functioned as a transcription activator causing activation of the NOTCH1 signaling pathway. Specific lncRNAs and their effects in T-ALL pathogenesis are summarized in Table 1.

4.3. LncRNAs as Cooperating Events of Genetic Abnormalities in B-ALL Pathogenesis

Fusion genes produced by chromosomal translocations are often strong oncogenic drivers and cytogenetic abnormalities in hematological malignancies such as acute myeloid leukemia (AML) or lymphoma [34,138]. Similarly, fusion oncogenes represent a prominent class of oncogenic drivers in ALL as well [8,9,139]. ETV6-RUNX1 generated by t(12;21) is the most common oncogenic fusion gene in childhood B-ALL [140]. A comprehensive analysis of the lncRNA transcriptome in ETV6-RUNX1+ B-ALL revealed the fusion protein-specific lncRNA expression signature [141]. Further analysis showed that expression of lncRNAs NKX2-3-1, TIMM21-5, ASTN1-1, and RTN4R-1 are linked to the oncogenic fusion protein. Knockdown of NKX2-3-1 and RTN4R-1 in ETV6-RUNX1+ cells reversed the expression of genes deregulated by the ETV6-RUNX1 fusion protein [141]. Likewise, lncRNA CASC15 is overexpressed in ETV6-RUNX1+ B-ALL, and shows increased expression of its chromosomally adjacent gene, SOX4, which encodes a transcriptional activator in lymphocytes, thereby upregulating cell survival, and proliferation [96]. An oncogenic lncRNA TCL6 was also upregulated in the ETV6-RUNX1+ B-ALL and probably associates with poor disease-free survival [123]. Collectively, these findings suggest the involvement of lncRNAs in fusion oncogene-mediated pathogenesis and progression of ALL.
Fusion of the mixed lineage leukemia (MLL) gene with other partner genes are frequent in pediatric leukemias, and MLL-rearranged B-ALL has long been considered a refractory disease due to the poor prognosis of associated patients [142]. A genome-wide lncRNA expression study found 52 upregulated and 59 downregulated lncRNAs between MLL-rearranged and MLL-unarranged ALL patient samples [143]; bioinformatics analysis showed that several lncRNAs correspond to the expression of the MLL fusion protein partner genes, such as HOXA and MEIS1 among others, and some other lncRNAs are associated with histone-related functions or membrane proteins; further, three subtypes of MLL rearranged ALL (MLL-AF4+, MLL-AF9+, and MLL-ENL+) showed a translocation specific lncNRA expression signature [143]. Another study reported that lncRNA BALR-6 is highly expressed in MLL-rearranged patient samples [98]; knockdown of BALR-6 reduced cell proliferation and induced apoptosis, while overexpression of BALR-6 caused a significant increase in early hematopoietic progenitor populations in murine BM transplantation experiments, suggesting that its dysregulation may cause developmental changes. Based on thorough literature searching, specific lncRNAs and their effects in B-ALL pathogenesis are summarized in Table 2.

4.4. LncRNAs and Immune Response Modulation in ALL Pathogenesis and Treatment

The immune suppressive tumor microenvironment is proven essential in the pathogenesis of many cancer types [144,145,146]. LncRNAs NONHSAT027612.2 and NONHSAT134556.2 were significantly elevated in the blood and BM of pediatric ALL patients, and may serve important roles in the pathogenesis of childhood ALL via suppressing immune response-associated pathways [125]. Besides, in the BM of patients with childhood T-ALL, membrane-localized and cytoplasm-localized lncRNA insulin receptor precursor (INSR) promoted CD4+ regulatory T-cell distribution and decreased the percentage of CD8+ cytotoxic T-cells in the BM of pediatric T-ALL patients, facilitating leukemic cell growth via immune suppression [147]. Mechanistically, through direct binding with INSR protein, lncRNA-INSR blocked the INSR ubiquitination site, causing abnormal accumulation and activation of INSR and the PI3K/AKT-signaling pathway. LncRNAs are also implicated in CART therapy of B-ALL [148]; specifically, a set of lncRNAs showed high degree of co-expression with transcription factors or histones (i.e., FOS and HIST1H4B) and were associated with immune processes during CAR-T therapy of B-ALL. These findings suggest that lncRNAs are important players in the immune response regulation in ALL pathogenesis and treatment.

4.5. LncRNAs in Susceptibility, Treatment Response and Prognosis of ALL

Single nucleotide polymorphism (SNP) is common in the human genome, and has profound implications in the pathogenesis/susceptibility of varying diseases. It was reported that the rs2147578 polymorphism of lncRNA LAMC2-1 may be a risk factor for developing childhood ALL, suggesting that SNPs might confer oncogenic properties on lncRNAs [149]. LncRNA PAX8-AS1 is located in the upstream region of PAX8, a gene encodes the transcription factor PAX8 required for cell growth and differentiation during embryonic development, and potentially regulates PAX8 expression [150]. Similarly, Bahari et al. reported that polymorphisms rs4848320 and rs6726151 of PAX8-AS1 might be risk factors for the development of childhood ALL [151]. Similarly, rs7158663 AG/AA genotypes of lncRNA MEG3 were associated with higher susceptibility to childhood ALL [152]. These findings indicate that some SNPs on lncRNAs are indicative of higher susceptibility for developing ALL.
Fernando et al. reported a correlation of high lncRNA BALR-2 expression with the diminished response to prednisone treatment and poor survival in patients [103]. BALR-2 knockdown led to reduced proliferation, increased apoptosis, as well as augmented sensitivity to prednisone treatment via activation of the glucocorticoid (GC) response pathway in both human and mouse B cells [103]. Another study reported that five lncRNAs are specifically upregulated in childhood B-ALL, and these lncRNAs had significant impacts on cell proliferation, migration, apoptosis, and treatment response [95]. In particular, expression of lncRNA RP11-137H2.4 was negatively associated with GC response [95]. Similarly, lncRNA GAS5 was also found to be a potential marker of GC response in remission induction therapy of childhood ALL [153]. A high expression of the lncRNA CDKN2B-AS1 level was associated with Adriamycin resistance [83]. Gioia et al. reported that LncRNAs RP11-624C23.1 and RP11-203E8 were downregulated in ALL, and restoring their expression in ALL cells’ increased sensitivity to genotoxic stress (e.g., chemotherapy agents), possibly by modulating the DNA damage response pathway [154]. These studies suggest that lncRNAs are important indicators of treatment response in ALL therapy.
Due to the strong oncogenic roles, some lncRNAs might predict prognosis in ALL patients. For example, lncRNA ZEB1-AS1 promoted the activation of IL-11/STAT3 signaling pathway by interacting with IL-11 in B-ALL cells, and a high expression of lncRNA ZEB1-AS1 predicted a poor prognosis for B-ALL patients [96]. A bioinformatics analysis presented a lncRNA-mRNA-based classifier that might be clinically useful to predict the recurrence and prognosis for childhood ALL [155]. It is also reported that the lncRNAs NEAT1 and MALAT1 sponges miR-335-3p, increasing the multidrug-resistance gene ATP-binding cassette sub-family A member 3 (ABCA3) expression and leading to poor prognosis in childhood ALL [156]. Many other studies also found similar roles of lncRNAs in prediction of the prognosis in ALL patients [78,106,113,140,151,157,158,159].
In conclusion, alterations in the expression of lncRNAs are important cooperating events in ALL pathogenesis, maintenance, and progression. Therefore, lncRNAs could be applied in diagnosis, classification, risk stratification, prognosis, as well as treatment of ALL.

5. Biogenesis and Biological Functions of Circular RNAs

Circular RNAs (circRNAs) represent a class of emerging ncRNA type, which are discovered by non-polyadenylated RNA-seq [50,160]. Similar to the lncRNAs, circRNAs are also transcribed by Pol II and have a wide range of size (100–10,000 nt) [160]. CircRNAs are the circularization products from precursor RNA splicing events, which can be generated from exons, introns, intergenic regions, untranslated regions (UTRs), or even tRNAs [160,161]. Generally, two major groups of circRNAs are generated from the Pol II-transcribed RNA precursors following splicing events [44,152,153,154,155,156,157,158,159,160,161,162,163,164]. On the one hand, back-splicing events of exons lead to connection of a downstream 5′ splice site with an upstream 3′ splice site to yield a group of circRNAs (Figure 3A) [162,163]. Orientation-opposite complementary sequences (OOCS), that juxtapose flanking introns of circularized exons to form RNA pairs or RNA binding proteins (RBPs) that bind to pre-mRNAs to connect flanking introns, generally facilitate back-splicing and circRNAs formation (Figure 3A) [162,163]. On the other hand, another group of circRNAs are generated from spliced intron lariats (Figure 3B) [164]; specifically, when the spliced intron lariats fail to debranch, covalent 2′,5′-phosphodiester bonds form to circularize these lariats, thereby generating circRNAs (known as intronic circRNAs) (Figure 3B) [164]. Accumulating evidence indicated that intronic circRNA formation depended on a consensus RNA motif containing a 7-nt GU-rich element near the 5′ splice site and an 11-nt C-rich element near the branch point (Figure 3B) [44,164]. Accordingly, circRNAs are characterized by a covalently closed loop structure, lacking both 5′- and 3′-ends, polyadenylated tail, and a 5′-CAP [160,161,162,163,164]. This unique structure confers exonuclease digestion resistance to circRNAs, largely enhancing their stability.
In comparison with mRNAs, circRNA production from pre-mRNA back-splicing is relatively slow, leading to a lower production rate of circRNAs than their linear mRNA counterparts [165]. CircRNAs containing one or more exons are exported to the cytoplasm [166]. These cytoplasmic circRNAs exert various biological functions such as miRNA sponges to regulate gene expression, protein complex coordinators, and others (Figure 3A). Meanwhile, the intronic circRNAs are retained in the nucleus and mainly regulate Pol II-mediated transcription (Figure 3B). In addition, accumulating evidence also indicated that some cytoplasmic circRNAs are transcribed into small peptides, thereby regulating protein–protein interactions [167]. Interestingly, the detected levels of some circRNAs are higher than their counterpart mRNAs, possibly due to their resistance to RNase clearance and intracellular accumulation [168]. The high stability of circRNAs also make them ideal biomarkers for liquid biopsy [169]. Collectively, circRNAs are important epigenetic players with various biological functions.
CircRNAs are implicated in cellular differentiation and maintenance of stem cell pluripotency [170,171]. For instance, a study found that the expression of circRNAs is cell type-specific and increases upon differentiation [170]. CircBIRC6 sponges inhibitory miRNAs such as miR-34a and miR-145, thereby activates multiple stemness markers including NANOG, OCT4, and SOX2, maintaining stem cell pluripotency [171]. These findings suggest that circRNAs might have pathological roles in cancer. Indeed, a large body of evidence demonstrated that circRNAs regulate hallmark phenotypes of cancer, such as angiogenesis, immune suppression, proliferation, cell cycle progression, apoptosis, invasion, and metastasis among others in many cancer types [161,172]. Therefore, circRNAs also represent a group of ncRNAs that is targetable in the fight against cancer.

6. CircRNAs in ALL Classification, Pathogenesis and Treatment

Owing to the unique circular structure, circRNAs display higher stability than other ncRNA types [169]. Similar to their role in other malignancies, circRNAs are also implicated in ALL classification, pathogenesis, and treatment, which will be recapitulated in detail in this part.

6.1. CircRNAs in ALL Classification and Diagnosis

Precise classification of ALL is instrumental for risk stratification and selection of optimal treatment strategies [8,9,10,173,174]. Thus, uncovering the circRNA signature of cytogenetic ALL subtypes is of critical importance in improving treatment outcomes. A study revealed specific circRNAs in B-cell (e.g., circPAX5, circAFF3, circIL4R, and circSETBP1) and T-cell (e.g., circIKZF1, circTNIK, circTXK, and circFBXW7) populations [175]. They also found upregulation of circPAX5, circPVT1, and circHIPK3 in pediatric B-ALL, and disclosed circRNAs with variable expression patterns across different cytogenetic subtypes of B-ALL; specifically, circAFF2 expression level was high in TCF3-PBX1+ B-ALL and, to a lesser extent, in ETV6-RUNX1+ B-ALL; intronic circBCL2 was upregulated in ETV6-RUNX1+ ALL; circSETBP1 and intergenic circX were both largely reduced in MLL rearranged B-ALL; circIKZF1 was lower in BCR-ABL+ and hyperdiploid B-ALL than the ETV6-RUNX1+ subtype which had similar expression with normal B-cells [175]. Likewise, another study investigated the circRNA signature of T-ALL [175]. According to their results, circZNF609, circKPNA5, circPSEN1, and circCEP70 were upregulated in ETP-ALL; circTASP1, circZBTB44, and circBACH1 were upregulated in TLX3-rearranged T-ALL; circHACD1, and circSTAM were upregulated in HOXA-rearranged T-ALL; circCAMSAP1 was upregulated in TLX1-rearranged T-ALL; and circCASC15 was upregulated in TAL/LMO subtype of T-ALL [176]. In addition, differences in the whole circRNAs’ expression signature, as well as specific circRNA expression, reportedly differentiated ALL from AML [177]. Collectively, these findings suggest that circRNAs could be applied in the classification, risk stratification, and even diagnosis of ALL.

6.2. CircRNAs Acting as Oncogenes in ALL Pathogenesis

Circular RNA PVT1 (circPVT1) was first identified as a suppressor of cellular senescence [178]. It was also demonstrated to be a proliferative factor and prognostic marker in gastric cancer [179]. Subsequently, Hu et al. reported that circPVT1 is also highly expressed in ALL patient samples and cell lines [180]; knockdown of circPVT1 in ALL cells resulted in proliferation inhibition and apoptosis induction via suppression of its neighboring gene c-Myc, and antiapoptotic Bcl-2 protein expression, suggesting an oncogenic role for this circRNA in ALL. Upregulation and oncogenic properties of circPVT1 in ALL were validated in other studies as well. For instance, the circPVT1 activated NF-κB signaling pathway through reducing miR-125b, thereby promoting ALL cell proliferation and migration [181]; this group also validated that circPVT1 sequesters the tumor suppressing miR-30e to increase DLL4 expression, thereby activating NOTCH signaling and promoting T-ALL cell proliferation [182]. In addition, it was demonstrated that upregulation of circ-0000745 in ALL cell lines could enhance the cell viability through the activation of ERK pathway [183]. According to another study, circ-0000745 was overexpressed in T-ALL patient BM and/or T-ALL cell lines, and circ-0000745 predominantly regulated NOTCH1 expression via sponging miR-193b-3p, contributing to the disease pathogenesis [184]. A recent study also demonstrated that circ-0000745 contributes to ALL development partly by sponging miR-494-3p to induce NET1 expression [185]. The oncogenic circPRKCI was upregulated in T-ALL patient samples and sequestered miR-20a-5p via sponging, thereby promoting viability and inhibiting apoptosis of T-ALL cells [186]. Circ-PRKDC was also upregulated in T-ALL patient samples as well as cell lines, and sponged miR-653-5p to depress RELN expression, leading to activation of the PI3K/AKT/mTOR signaling pathway and promotion of cell proliferation, as well as inhibition of apoptosis [187]. Collectively, these findings suggest that certain oncogenic circRNAs are upregulated in ALL and contribute to the pathogenesis of disease, and these circRNAs are potential therapeutic targets for the diagnosis and treatment of ALL.

6.3. CircRNAs Acting as Tumor Suppressors in ALL Pathogenesis

On the other hand, circRNAs also exert tumor suppressor functions. It was reported that the cytoplasm-localized circ_0000094 expression is markedly reduced in T-ALL [188]; intriguingly, exogenous compensation of circ_0000094 inhibited T-ALL cell proliferation, migration, and invasion, and enhanced apoptosis; while knockdown of circ_0000094 showed the opposite effect. Further analysis revealed that circ_0000094 is a molecular sponge for miR-223-3p, and it could up-regulate the expression of tumor suppressor FBW7 via sequestering miR-223-3p [188]. Expression of circ_0000143 was also significantly reduced in T-ALL patient samples compared to healthy controls; overexpression of circ_0000143 inhibited T-ALL cell viability, migration and invasion, and induced apoptosis. Mechanistically, circ_0000143 acted as a molecular sponge to sequester miR-142-3p, thereby upregulating miR142-3p repressed glucocorticoid receptor α (GRα) expression [189]. Likewise, circADD2 was downregulated in ALL patient samples and cell lines [190]; overexpression of circADD2 inhibited cell proliferation and promoted apoptosis; while, knockdown of circADD2 showed the opposite effect; mechanistically, circADD2 could directly sponge miR-149-5p, a positive regulator of AKT2, leading to reduction of AKT2 and pAKT2 level [190]. These findings suggest that tumor suppressing circRNAs are downregulated in ALL, and increasing expression of these circRNAs represents a novel strategy in ALL treatment as well.
Taken together, circRNAs play carcinogenic or anticancer effects in ALL pathogenesis and progression. Their high stability could provide a unique advantage in liquid biopsy-based diagnosis. More importantly, RNA interference-based strategies or manipulation of endogenous promoters by CRISPR/Cas9-based strategies are feasible approaches to decrease oncogenic or increase tumor suppressing circRNAs expression [191]. Thus, circRNAs could be applied in diagnosis, classification, and treatment of ALL.

7. Conclusions

Over the last decade, tremendous progress has been made in the treatment of ALL using evidence-based, risk-adapted, MRD-guided, and multiagent chemotherapy [8,9,10,29]. Nevertheless, disease relapse still occurs for many adult patients [10,30,39]. Personalized medicine targeting the initiation, maintenance, progression, and resistance mechanisms is a promising strategy to increase survival in this set of patients [17,39]. However, although most of the genetic abnormalities resulting in ALL onset and progression are clearly defined, limited targeted therapeutics are available in clinic [10,18,37,192]. This is probably due to the low translational value of some therapeutic targets or unexpected adverse events. Thus, there is an urgent need to excavate novel therapeutic targets. As important cooperating events of genetic changes in ALL etiology, maintenance, and progression, lncRNAs and circRNAs are proven as novel classes of targetable vulnerabilities in ALL. Especially for those lnc/circRNAs specifically upregulated in certain ALL subtypes, it is facile to design and produce siRNAs or ASOs or CRISPR/Cas based tools that target and downregulate their level [190]. These comprehensive analysis and insights could facilitate the bridging of basic research to the clinical practice, promoting emergence of new therapeutic strategies to improve ALL patient outcomes.

Author Contributions

Conceptualization, Y.M.; writing—original draft preparation, Y.M., T.Y. and L.Y.; writing—review and editing, Y.M., H.N. and W.Y.; supervision, H.N. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Zhejiang Provincial Natural Science Foundation of China (LD22H310005), the National Natural Science Foundation of China (No. 82170143, 82000155, 81872942), Zhejiang Innovation Team Grant (2020R01006), and the China Postdoctoral Science Foundation (No. 2021M702877).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ALLacute lymphoblastic leukemia
Allo-SCTallogenic stem cell transplantation
ASOsantisense oligonucleotides
BMbone marrow
CARTchimeric antigen T-cell therapy
CNScentral nervous system
CRcomplete remission
circRNAcircular RNA
DFSdisease-free survival
ENCODEencyclopedia of DNA elements
ETP-ALLearly T-cell precursor ALL
GCglucocorticoid
lncRNAlong non-coding RNA
MRDminimal residual disease
MLLmixed lineage leukemia gene
ncRNAnon-coding RNA
ntnucleotide
OOCSorientation-opposite complementary sequences
ORFsopen reading frames
Pol IIRNA polymerase II
PRCpolycome repressive complex
RBPsRNA binding proteins
RNA-seqRNA-sequencing
SNPSingle nucleotide polymorphism
TKIstyrosine kinase inhibitors
TAL1T-cell acute lymphocytic leukemia protein 1
TLX1T-Cell Leukemia Homeobox 1

References

  1. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [Green Version]
  2. Saw, P.E.; Xu, X.; Chen, J.; Song, E.W. Non-coding RNAs: The new central dogma of cancer biology. Sci. China Life Sci. 2021, 64, 22–50. [Google Scholar] [CrossRef] [PubMed]
  3. Bindereif, A. Pre-mRNA splicing: From protein-coding to noncoding RNAs. RNA 2015, 21, 571–572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 2012, 489, 57–74. [Google Scholar] [CrossRef]
  5. Djebali, S.; Davis, C.A.; Merkel, A.; Dobin, A.; Lassmann, T.; Mortazavi, A.; Tanzer, A.; Lagarde, J.; Lin, W.; Schlesinger, F.; et al. Landscape of transcription in human cells. Nature 2012, 489, 101–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Hombach, S.; Kretz, M. Non-coding RNAs: Classification, Biology and Functioning. Adv. Exp. Med. Biol. 2016, 937, 3–17. [Google Scholar] [PubMed]
  7. Zhang, P.; Wu, W.; Chen, Q.; Chen, M. Non-Coding RNAs and their Integrated Networks. J. Integr. Bioinform. 2019, 16, 20190027. [Google Scholar] [CrossRef]
  8. Malard, F.; Mohty, M. Acute lymphoblastic leukaemia. Lancet 2020, 395, 1146–1162. [Google Scholar] [CrossRef]
  9. Terwilliger, T.; Abdul-Hay, M. Acute lymphoblastic leukemia: A comprehensive review and 2017 update. Blood Cancer J. 2017, 7, e577. [Google Scholar] [CrossRef] [Green Version]
  10. van der Valentina Cordo, J.C.; Zwet, K.C.; Rob Pieters, J.P. T-cell Acute Lymphoblastic Leukemia: A Roadmap to Targeted Therapies. Blood Cancer Discov. 2020, 2, 19–31. [Google Scholar] [CrossRef]
  11. Ratti, S.; Lonetti, A.; Follo, M.Y.; Paganelli, F.; Martelli, A.M.; Chiarini, F.; Evangelisti, C. B-ALL Complexity: Is Targeted Therapy Still A Valuable Approach for Pediatric Patients? Cancers 2020, 12, 3498. [Google Scholar] [CrossRef] [PubMed]
  12. Fischer, U.; Yang, J.J.; Ikawa, T.; Hein, D.; Vicente-Dueñas, C.; Borkhardt, A.; Sánchez-García, I. Cell Fate Decisions: The Role of Transcription Factors in Early B-cell Development and Leukemia. Blood Cancer Discov. 2020, 1, 224–233. [Google Scholar] [CrossRef] [PubMed]
  13. Van der Zwet, J.C.G.; Cordo, V.; Canté-Barrett, K.; Meijerink, J.P.P. Multi-omic approaches to improve outcome for T-cell acute lymphoblastic leukemia patients. Adv. Biol. Regul. 2019, 74, 100647. [Google Scholar] [CrossRef] [PubMed]
  14. Ogawa, S. Deciphering the Clonal Origin of Relapsed Acute Lymphoblastic Leukemia in Children. Blood Cancer Discov. 2020, 1, 21–22. [Google Scholar] [CrossRef]
  15. Lee, S.H.R.; Li, Z.; Tai, S.T.; Oh, B.L.Z.; Yeoh, A.E.J. Genetic Alterations in Childhood Acute Lymphoblastic Leukemia: Interactions with Clinical Features and Treatment Response. Cancers 2021, 13, 4068. [Google Scholar] [CrossRef] [PubMed]
  16. Paul, S.; Kantarjian, H.; Jabbour, E.J. Adult Acute Lymphoblastic Leukemia. Mayo Clin. Proc. 2016, 91, 1645–1666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Pui, C.H. Precision medicine in acute lymphoblastic leukemia. Front. Med. 2020, 14, 689–700. [Google Scholar] [CrossRef]
  18. Waanders, E.; Gu, Z.; Dobson, S.M.; Antić, Ž.; Crawford, J.C.; Ma, X.; Edmonson, M.N.; Payne-Turner, D.; van de Vorst, M.; Jongmans, M.; et al. Mutational landscape and patterns of clonal evolution in relapsed pediatric acute lymphoblastic leukemia. Blood Cancer Discov. 2020, 1, 96–111. [Google Scholar] [CrossRef]
  19. Wiemels, J.L.; Cazzaniga, G.; Daniotti, M.; Eden, O.B.; Addison, G.M.; Masera, G.; Saha, V.; Biondi, A.; Greaves, M.F. Prenatal origin of acute lymphoblastic leukaemia in children. Lancet 1999, 354, 1499–1503. [Google Scholar] [CrossRef]
  20. Andreasson, P.; Schwaller, J.; Anastasiadou, E.; Aster, J.; Gilliland, D.G. The expression of ETV6/CBFA2 (TEL/AML1) is not sufficient for the transformation of hematopoietic cell lines in vitro or the induction of hematologic disease in vivo. Cancer Genet. Cytogenet. 2001, 130, 93–104. [Google Scholar] [CrossRef]
  21. Rodriguez, P.D.; Paculova, H.; Kogut, S.; Heath, J.; Schjerven, H.; Frietze, S. Non-Coding RNA Signatures of B-Cell Acute Lymphoblastic Leukemia. Int. J. Mol. Sci. 2021, 22, 2683. [Google Scholar] [CrossRef] [PubMed]
  22. Deng, W.; Pan, M.; Zhu, S.; Chao, R.; Wang, L. Emerging roles of microRNAs in acute lymphoblastic leukemia and their clinical prospects. Expert Rev. Hematol. 2021, 14, 987–992. [Google Scholar] [CrossRef] [PubMed]
  23. Grobbelaar, C.; Ford, A.M. The Role of MicroRNA in Paediatric Acute Lymphoblastic Leukaemia: Challenges for Diagnosis and Therapy. J. Oncol. 2019, 2019, 8941471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Malagola, M.; Papayannidis, C.; Baccarani, M. Tyrosine kinase inhibitors in Ph+ acute lymphoblastic leukaemia: Facts and perspectives. Ann. Hematol. 2016, 95, 681–693. [Google Scholar] [CrossRef]
  25. Hangai, M.; Urayama, K.Y.; Tanaka, J.; Kato, K.; Nishiwaki, S.; Koh, K.; Noguchi, M.; Kato, K.; Yoshida, N.; Sato, M.; et al. Allogeneic Stem Cell Transplantation for Acute Lymphoblastic Leukemia in Adolescents and Young Adults. Biol. Blood Marrow Transplant. 2019, 25, 1597–1602. [Google Scholar] [CrossRef]
  26. Lato, M.W.; Przysucha, A.; Grosman, S.; Zawitkowska, J.; Lejman, M. The New Therapeutic Strategies in Pediatric T-Cell Acute Lymphoblastic Leukemia. Int. J. Mol. Sci. 2021, 22, 94502. [Google Scholar] [CrossRef]
  27. Zheng, S.; Gillespie, E.; Naqvi, A.S.; Hayer, K.E.; Ang, Z.; Torres-Diz, M.; Quesnel-Vallières, M.; Hottman, D.A.; Bagashev, A.; Chukinas, J.; et al. Modulation of CD22 Protein Expression in Childhood Leukemia by Pervasive Splicing Aberrations: Implications for CD22-Directed Immunotherapies. Blood Cancer Discov. 2022, 3, 103–115. [Google Scholar] [CrossRef]
  28. Bartram, J.; Veys, P.; Vora, A. Improvements in outcome of childhood acute lymphoblastic leukaemia (ALL) in the UK—A success story of modern medicine through successive UKALL trials and international collaboration. Br. J. Haematol. 2020, 191, 562–567. [Google Scholar]
  29. Pulsipher, M.A.; Han, X.; Maude, S.L.; Laetsch, T.W.; Qayed, M.; Rives, S.; Boyer, M.W.; Hiramatsu, H.; Yanik, G.A.; Driscoll, T.; et al. Next-Generation Sequencing of Minimal Residual Disease for Predicting Relapse after Tisagenlecleucel in Children and Young Adults with Acute Lymphoblastic Leukemia. Blood Cancer Discov. 2022, 3, 66–81. [Google Scholar] [CrossRef]
  30. Ronson, A.; Tvito, A.; Rowe, J.M. Treatment of Relapsed/Refractory Acute Lymphoblastic Leukemia in Adults. Curr. Oncol. Rep. 2016, 18, 39. [Google Scholar] [CrossRef]
  31. Tottone, L.; Lancho, O.; Loh, J.W.; Singh, A.; Kimura, S.; Roels, J.; Kuchmiy, A.; Strubbe, S.; Lawlor, M.A.; da Silva-Diz, V.; et al. A Tumor Suppressor Enhancer of PTEN in T-cell development and leukemia. Blood Cancer Discov. 2021, 2, 92–109. [Google Scholar] [CrossRef] [PubMed]
  32. Fielding, A.K.; Rowe, J.M.; Buck, G.; Foroni, L.; Gerrard, G.; Litzow, M.R.; Lazarus, H.; Luger, S.M.; Marks, D.I.; McMillan, A.K.; et al. UKALLXII/ECOG2993: Addition of imatinib to a standard treatment regimen enhances long-term outcomes in Philadelphia positive acute lymphoblastic leukemia. Blood 2014, 123, 843–850. [Google Scholar] [CrossRef] [PubMed]
  33. Foà, R.; Bassan, R.; Vitale, A.; Elia, L.; Piciocchi, A.; Puzzolo, M.C.; Canichella, M.; Viero, P.; Ferrara, F.; Lunghi, M.; et al. Dasatinib-Blinatumomab for Ph-Positive Acute Lymphoblastic Leukemia in Adults. N. Engl. J. Med. 2020, 383, 1613–1623. [Google Scholar] [CrossRef] [PubMed]
  34. Maimaitiyiming, Y.; Wang, Q.Q.; Yang, C.; Ogra, Y.; Lou, Y.; Smith, C.A.; Hussain, L.; Shao, Y.M.; Lin, J.; Liu, J.; et al. Hyperthermia Selectively Destabilizes Oncogenic Fusion Proteins. Blood Cancer Discov. 2021, 2, 388–401. [Google Scholar] [CrossRef]
  35. Jin, M.; An, Q.; Xu, S. Central nervous system disease in childhood acute lymphoblastic leukemia. Minerva Pediatr. 2019, 71, 470–474. [Google Scholar] [CrossRef]
  36. Simone, J.V. Preventive central-nervous-system therapy in acute leukemia. N. Engl. J. Med. 1973, 289, 1248–1249. [Google Scholar] [CrossRef]
  37. Pui, C.H.; Howard, S.C. Current management and challenges of malignant disease in the CNS in paediatric leukaemia. Lancet Oncol. 2008, 9, 257–268. [Google Scholar] [CrossRef]
  38. Frishman-Levy, L.; Izraeli, S. Advances in understanding the pathogenesis of CNS acute lymphoblastic leukaemia and potential for therapy. Br. J. Haematol. 2017, 176, 157–167. [Google Scholar] [CrossRef]
  39. Bhojwani, D.; Pui, C.H. Relapsed childhood acute lymphoblastic leukaemia. Lancet Oncol. 2013, 14, e205–e217. [Google Scholar] [CrossRef]
  40. Barredo, J.C.; Devidas, M.; Lauer, S.J.; Billett, A.; Marymont, M.; Pullen, J.; Camitta, B.; Winick, N.; Carroll, W.; Ritchey, A.K. Isolated CNS relapse of acute lymphoblastic leukemia treated with intensive systemic chemotherapy and delayed CNS radiation: A pediatric oncology group study. J. Clin. Oncol. 2006, 24, 3142–3149. [Google Scholar] [CrossRef]
  41. Lenk, L.; Alsadeq, A.; Schewe, D.M. Involvement of the central nervous system in acute lymphoblastic leukemia: Opinions on molecular mechanisms and clinical implications based on recent data. Cancer Metastasis Rev. 2020, 39, 173–187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Segers, H.; Cools, J. Genomics, Transcriptomics, and Minimal Residual Disease Detection: The Winning Team to Guide Treatment of Acute Lymphoblastic Leukemia. Blood Cancer Discov. 2021, 2, 294–296. [Google Scholar] [CrossRef] [PubMed]
  43. Vanner, R.J.; Dobson, S.M.; Gan, O.I.; McLeod, J.; Schoof, E.M.; Grandal, I.; Wintersinger, J.A.; Garcia-Prat, L.; Hosseini, M.; Xie, S.Z.; et al. Multiomic Profiling of Central Nervous System Leukemia Identifies mRNA Translation as a Therapeutic Target. Blood Cancer Discov. 2022, 3, 16–31. [Google Scholar] [CrossRef] [PubMed]
  44. Wu, H.; Yang, L.; Chen, L.L. The Diversity of Long Noncoding RNAs and Their Generation. Trends Genet. 2017, 33, 540–552. [Google Scholar] [CrossRef] [PubMed]
  45. Chen, D.; Lu, T.; Tan, J.; Li, H.; Wang, Q.; Wei, L. Long Non-coding RNAs as Communicators and Mediators Between the Tumor Microenvironment and Cancer Cells. Front. Oncol. 2019, 9, 739. [Google Scholar] [CrossRef] [PubMed]
  46. Marahrens, Y.; Panning, B.; Dausman, J.; Strauss, W.; Jaenisch, R. Xist-deficient mice are defective in dosage compensation but not spermatogenesis. Genes Dev. 1997, 11, 156–166. [Google Scholar] [CrossRef] [Green Version]
  47. Xing, Y.H.; Yao, R.W.; Zhang, Y.; Guo, C.J.; Jiang, S.; Xu, G.; Dong, R.; Yang, L.; Chen, L.L. SLERT Regulates DDX21 Rings Associated with Pol I Transcription. Cell 2017, 169, 664–678.e16. [Google Scholar] [CrossRef]
  48. Statello, L.; Guo, C.J.; Chen, L.L.; Huarte, M. Gene regulation by long non-coding RNAs and its biological functions. Nat. Rev. Mol. Cell Biol. 2021, 22, 96–118. [Google Scholar] [CrossRef]
  49. Wilusz, J.E.; Freier, S.M.; Spector, D.L. 3′ end processing of a long nuclear-retained noncoding RNA yields a tRNA-like cytoplasmic RNA. Cell 2008, 135, 919–932. [Google Scholar] [CrossRef] [Green Version]
  50. Yin, Q.F.; Yang, L.; Zhang, Y.; Xiang, J.F.; Wu, Y.W.; Carmichael, G.G.; Chen, L.L. Long noncoding RNAs with snoRNA ends. Mol. Cell 2012, 48, 219–230. [Google Scholar] [CrossRef] [Green Version]
  51. Wu, H.; Yin, Q.F.; Luo, Z.; Yao, R.W.; Zheng, C.C.; Zhang, J.; Xiang, J.F.; Yang, L.; Chen, L.L. Unusual Processing Generates SPA LncRNAs that Sequester Multiple RNA Binding Proteins. Mol. Cell 2016, 64, 534–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Holdt, L.M.; Hoffmann, S.; Sass, K.; Langenberger, D.; Scholz, M.; Krohn, K.; Finstermeier, K.; Stahringer, A.; Wilfert, W.; Beutner, F.; et al. Alu elements in ANRIL non-coding RNA at chromosome 9p21 modulate atherogenic cell functions through trans-regulation of gene networks. PLoS Genet. 2013, 9, e1003588. [Google Scholar] [CrossRef] [PubMed]
  53. Yap, K.L.; Li, S.; Muñoz-Cabello, A.M.; Raguz, S.; Zeng, L.; Mujtaba, S.; Gil, J.; Walsh, M.J.; Zhou, M.M. Molecular interplay of the noncoding RNA ANRIL and methylated histone H3 lysine 27 by polycomb CBX7 in transcriptional silencing of INK4a. Mol. Cell 2010, 38, 662–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Jeon, Y.; Lee, J.T. YY1 tethers Xist RNA to the inactive X nucleation center. Cell 2011, 146, 119–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. McHugh, C.A.; Chen, C.K.; Chow, A.; Surka, C.F.; Tran, C.; McDonel, P.; Pandya-Jones, A.; Blanco, M.; Burghard, C.; Moradian, A.; et al. The Xist lncRNA interacts directly with SHARP to silence transcription through HDAC3. Nature 2015, 521, 232–236. [Google Scholar] [CrossRef] [PubMed]
  56. Colognori, D.; Sunwoo, H.; Kriz, A.J.; Wang, C.Y.; Lee, J.T. Xist Deletional Analysis Reveals an Interdependency between Xist RNA and Polycomb Complexes for Spreading along the Inactive, X. Mol. Cell 2019, 74, 101–117.e10. [Google Scholar] [CrossRef] [Green Version]
  57. Schlackow, M.; Nojima, T.; Gomes, T.; Dhir, A.; Carmo-Fonseca, M.; Proudfoot, N.J. Distinctive Patterns of Transcription and RNA Processing for Human lincRNAs. Mol. Cell 2017, 65, 25–38. [Google Scholar] [CrossRef] [Green Version]
  58. Zuckerman, B.; Ulitsky, I. Predictive models of subcellular localization of long RNAs. RNA 2019, 25, 557–572. [Google Scholar] [CrossRef]
  59. Yamazaki, T.; Souquere, S.; Chujo, T.; Kobelke, S.; Chong, Y.S.; Fox, A.H.; Bond, C.S.; Nakagawa, S.; Pierron, G.; Hirose, T. Functional Domains of NEAT1 Architectural lncRNA Induce Paraspeckle Assembly through Phase Separation. Mol. Cell 2018, 70, 1038–1053.e7. [Google Scholar] [CrossRef] [Green Version]
  60. Hutchinson, J.N.; Ensminger, A.W.; Clemson, C.M.; Lynch, C.R.; Lawrence, J.B.; Chess, A. A screen for nuclear transcripts identifies two linked noncoding RNAs associated with SC35 splicing domains. BMC Genom. 2007, 8, 39. [Google Scholar] [CrossRef] [Green Version]
  61. Engreitz, J.M.; Sirokman, K.; McDonel, P.; Shishkin, A.A.; Surka, C.; Russell, P.; Grossman, S.R.; Chow, A.Y.; Guttman, M.; Lander, E.S. RNA-RNA interactions enable specific targeting of noncoding RNAs to nascent Pre-mRNAs and chromatin sites. Cell 2014, 159, 188–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Maharana, S.; Wang, J.; Papadopoulos, D.K.; Richter, D.; Pozniakovsky, A.; Poser, I.; Bickle, M.; Rizk, S.; Guillén-Boixet, J.; Franzmann, T.M.; et al. RNA buffers the phase separation behavior of prion-like RNA binding proteins. Science 2018, 360, 918–921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Su, Y.; Maimaitiyiming, Y.; Wang, L.; Cheng, X.; Hsu, C.H. Modulation of Phase Separation by RNA: A Glimpse on N6-Methyladenosine Modification. Front. Cell Dev. Biol. 2021, 9, 786454. [Google Scholar] [CrossRef] [PubMed]
  64. Lagier-Tourenne, C.; Polymenidou, M.; Cleveland, D.W. TDP-43 and FUS/TLS: Emerging roles in RNA processing and neurodegeneration. Hum. Mol. Genet. 2010, 19, R46–R64. [Google Scholar] [CrossRef] [PubMed]
  65. Zuckerman, B.; Ron, M.; Mikl, M.; Segal, E.; Ulitsky, I. Gene Architecture and Sequence Composition Underpin Selective Dependency of Nuclear Export of Long RNAs on NXF1 and the TREX Complex. Mol. Cell 2020, 79, 251–267.e6. [Google Scholar] [CrossRef]
  66. Huarte, M. The emerging role of lncRNAs in cancer. Nat. Med. 2015, 21, 1253–1261. [Google Scholar] [CrossRef]
  67. Nandwani, A.; Rathore, S.; Datta, M. LncRNAs in cancer: Regulatory and therapeutic implications. Cancer Lett. 2021, 501, 162–171. [Google Scholar] [CrossRef]
  68. Melixetian, M.; Pelicci, P.G.; Lanfrancone, L. Regulation of LncRNAs in Melanoma and Their Functional Roles in the Metastatic Process. Cells 2022, 11, 577. [Google Scholar] [CrossRef]
  69. Nasrollahzadeh-Khakiani, M.; Emadi-Baygi, M.; Schulz, W.A.; Nikpour, P. Long noncoding RNAs in gastric cancer carcinogenesis and metastasis. Brief. Funct. Genom. 2017, 16, 129–145. [Google Scholar] [CrossRef]
  70. Liu, H.; Wan, J.; Chu, J. Long non-coding RNAs and endometrial cancer. Biomed. Pharmacother. 2019, 119, 109396. [Google Scholar] [CrossRef]
  71. Lee, J.S.; Mendell, J.T. Antisense-Mediated Transcript Knockdown Triggers Premature Transcription Termination. Mol. Cell 2020, 77, 1044–1054.e3. [Google Scholar] [CrossRef] [PubMed]
  72. Stojic, L.; Niemczyk, M.; Orjalo, A.; Ito, Y.; Ruijter, A.E.; Uribe-Lewis, S.; Joseph, N.; Weston, S.; Menon, S.; Odom, D.T.; et al. Transcriptional silencing of long noncoding RNA GNG12-AS1 uncouples its transcriptional and product-related functions. Nat. Commun. 2016, 7, 10406. [Google Scholar] [CrossRef] [PubMed]
  73. Zhang, X.; Lian, Z.; Padden, C.; Gerstein, M.B.; Rozowsky, J.; Snyder, M.; Gingeras, T.R.; Kapranov, P.; Weissman, S.M.; Newburger, P.E. A myelopoiesis-associated regulatory intergenic noncoding RNA transcript within the human HOXA cluster. Blood 2009, 113, 2526–2534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Ørom, U.A.; Derrien, T.; Beringer, M.; Gumireddy, K.; Gardini, A.; Bussotti, G.; Lai, F.; Zytnicki, M.; Notredame, C.; Huang, Q.; et al. Long noncoding RNAs with enhancer-like function in human cells. Cell 2010, 143, 46–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Xia, F.; Dong, F.; Yang, Y.; Huang, A.; Chen, S.; Sun, D.; Xiong, S.; Zhang, J. Dynamic transcription of long non-coding RNA genes during CD4+ T cell development and activation. PLoS ONE 2014, 9, e101588. [Google Scholar] [CrossRef] [Green Version]
  76. Brazão, T.F.; Johnson, J.S.; Müller, J.; Heger, A.; Ponting, C.P.; Tybulewicz, V.L. Long noncoding RNAs in B-cell development and activation. Blood 2016, 128, e10–e19. [Google Scholar] [CrossRef] [Green Version]
  77. Trimarchi, T.; Bilal, E.; Ntziachristos, P.; Fabbri, G.; Dalla-Favera, R.; Tsirigos, A.; Aifantis, I. Genome-wide mapping and characterization of Notch-regulated long noncoding RNAs in acute leukemia. Cell 2014, 158, 593–606. [Google Scholar] [CrossRef] [Green Version]
  78. El-Khazragy, N.; Abdel Aziz, M.A.; Hesham, M.; Matbouly, S.; Mostafa, S.A.; Bakkar, A.; Abouelnile, M.; Noufal, Y.; Mahran, N.A.; Elkhalek, M.A.; et al. Upregulation of leukemia-induced non-coding activator RNA (LUNAR1) predicts poor outcome in pediatric T-acute lymphoblastic leukemia. Immunobiology 2021, 226, 152149. [Google Scholar] [CrossRef]
  79. Zhang, L.; Xu, H.G.; Lu, C. A novel long non-coding RNA T-ALL-R-LncR1 knockdown and Par-4 cooperate to induce cellular apoptosis in T-cell acute lymphoblastic leukemia cells. Leuk. Lymphoma 2014, 55, 1373–1382. [Google Scholar] [CrossRef]
  80. Wang, Y.; Wu, P.; Lin, R.; Rong, L.; Xue, Y.; Fang, Y. LncRNA NALT interaction with NOTCH1 promoted cell proliferation in pediatric T cell acute lymphoblastic leukemia. Sci. Rep. 2015, 5, 13749. [Google Scholar] [CrossRef] [Green Version]
  81. Renou, L.; Boelle, P.Y.; Deswarte, C.; Spicuglia, S.; Benyoucef, A.; Calvo, J.; Uzan, B.; Belhocine, M.; Cieslak, A.; Landman-Parker, J.; et al. Homeobox protein TLX3 activates miR-125b expression to promote T-cell acute lymphoblastic leukemia. Blood Adv. 2017, 1, 733–747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Tan, S.H.; Leong, W.Z.; Ngoc, P.C.T.; Tan, T.K.; Bertulfo, F.C.; Lim, M.C.; An, O.; Li, Z.; Yeoh, A.; Fullwood, M.J.; et al. The enhancer RNA ARIEL activates the oncogenic transcriptional program in T-cell acute lymphoblastic leukemia. Blood 2019, 134, 239–251. [Google Scholar] [CrossRef] [PubMed]
  83. Chen, L.; Shi, Y.; Li, J.; Yang, X.; Li, R.; Zhou, X.; Zhu, L. LncRNA CDKN2B-AS1 contributes to tumorigenesis and chemoresistance in pediatric T-cell acute lymphoblastic leukemia through miR-335-3p/TRAF5 axis. Anticancer Drugs 2020. Epub ahead of print. [Google Scholar] [CrossRef] [PubMed]
  84. Li, S.; Guo, W.; Geng, H.; Wang, C.; Yang, S.; Xu, X. LINC00511 exacerbated T-cell acute lymphoblastic leukemia via miR-195-5p/LRRK1 axis. Biosci. Rep. 2020, 40, BSR20193631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Li, G.; Gao, L.; Zhao, J.; Liu, D.; Li, H.; Hu, M. LncRNA ANRIL/miR-7-5p/TCF4 axis contributes to the progression of T cell acute lymphoblastic leukemia. Cancer Cell Int. 2020, 20, 335. [Google Scholar] [CrossRef]
  86. Li, X.; Song, F.; Sun, H. Long non-coding RNA AWPPH interacts with ROCK2 and regulates the proliferation and apoptosis of cancer cells in pediatric T-cell acute lymphoblastic leukemia. Oncol. Lett. 2020, 20, 239. [Google Scholar] [CrossRef]
  87. Luo, Y.Y.; Wang, Z.H.; Yu, Q.; Yuan, L.L.; Peng, H.L.; Xu, Y.X. LncRNA-NEAT1 promotes proliferation of T-ALL cells via miR-146b-5p/NOTCH1 signaling pathway. Pathol. Res. Pract. 2020, 216, 153212. [Google Scholar] [CrossRef]
  88. Singh, N.; Padi, S.K.R.; Bearss, J.J.; Pandey, R.; Okumura, K.; Beltran, H.; Song, J.H.; Kraft, A.S.; Olive, V. PIM protein kinases regulate the level of the long noncoding RNA H19 to control stem cell gene transcription and modulate tumor growth. Mol. Oncol. 2020, 14, 974–990. [Google Scholar] [CrossRef] [Green Version]
  89. Mofidi, M.; Rahgozar, S.; Pouyanrad, S. Increased level of long non coding RNA H19 is correlated with the downregulation of miR-326 and BCL-2 genes in pediatric acute lymphoblastic leukemia, a possible hallmark for leukemogenesis. Mol. Biol. Rep. 2021, 48, 1531–1538. [Google Scholar] [CrossRef]
  90. Norouzi, A.; Motaghi, M.; Hassanshahi, G.; Nazari-Robati, M. Exploring the expression profile of vitamin D receptor and its related long non-coding RNAs in patients with acute lymphoblastic leukemia. Rev. Assoc. Med. Bras. 2021, 67, 1113–1117. [Google Scholar] [CrossRef]
  91. Li, J.; Muhammad, J.; Xie, T.; Sun, J.; Lei, Y.; Wei, Z.; Pan, S.; Qin, H.; Shao, L.; Jiang, D.; et al. LINC00853 restrains T cell acute lymphoblastic leukemia invasion and infiltration by regulating CCR9/CCL25. Mol. Immunol. 2021, 140, 267–275. [Google Scholar] [CrossRef]
  92. Xiao, S.; Xu, N.; Ding, Q.; Huang, S.; Zha, Y.; Zhu, H. LncRNA VPS9D1-AS1 promotes cell proliferation in acute lymphoblastic leukemia through modulating GPX1 expression by miR-491-5p and miR-214-3p evasion. Biosci. Rep. 2020, 40, BSR20193461. [Google Scholar] [CrossRef]
  93. Huang, M.; Zheng, J.; Ren, Y.; Zhu, J.; Kou, L.; Nie, J. LINC00221 suppresses the malignancy of children acute lymphoblastic leukemia. Biosci. Rep. 2020, 40, BSR20194070. [Google Scholar] [CrossRef] [Green Version]
  94. Ouimet, M.; Drouin, S.; Lajoie, M.; Caron, M.; St-Onge, P.; Gioia, R.; Richer, C.; Sinnett, D. A childhood acute lymphoblastic leukemia-specific lncRNA implicated in prednisolone resistance, cell proliferation, and migration. Oncotarget 2017, 8, 7477–7488. [Google Scholar] [CrossRef] [Green Version]
  95. Fernando, T.R.; Contreras, J.R.; Zampini, M.; Rodriguez-Malave, N.I.; Alberti, M.O.; Anguiano, J.; Tran, T.M.; Palanichamy, J.K.; Gajeton, J.; Ung, N.M.; et al. The lncRNA CASC15 regulates SOX4 expression in RUNX1-rearranged acute leukemia. Mol. Cancer. 2017, 16, 126. [Google Scholar] [CrossRef]
  96. Wang, Q.; Du, X.; Yang, M.; Xiao, S.; Cao, J.; Song, J.; Wang, L. LncRNA ZEB1-AS1 contributes to STAT3 activation by associating with IL-11 in B-lymphoblastic leukemia. Biotechnol. Lett. 2017, 39, 1801–1810. [Google Scholar] [CrossRef]
  97. Rodríguez-Malavé, N.I.; Fernando, T.R.; Patel, P.C.; Contreras, J.R.; Palanichamy, J.K.; Tran, T.M.; Anguiano, J.; Davoren, M.J.; Alberti, M.O.; Pioli, K.T.; et al. BALR-6 regulates cell growth and cell survival in B-lymphoblastic leukemia. Mol. Cancer 2015, 14, 214. [Google Scholar] [CrossRef] [Green Version]
  98. Wang, X.; Yang, J.; Guo, G.; Feng, R.; Chen, K.; Liao, Y.; Zhang, L.; Sun, L.; Huang, S.; Chen, J.L. Novel lncRNA-IUR suppresses Bcr-Abl-induced tumorigenesis through regulation of STAT5-CD71 pathway. Mol. Cancer 2019, 18, 84. [Google Scholar] [CrossRef]
  99. Wang, W.; Wu, F.; Ma, P.; Gan, S.; Li, X.; Chen, L.; Sun, L.; Sun, H.; Jiang, Z.; Guo, F. LncRNA CRNDE Promotes the Progression of B-cell Precursor Acute Lymphoblastic Leukemia by Targeting the miR-345-5p/CREB Axis. Mol. Cells 2020, 43, 718–727. [Google Scholar]
  100. Wang, W.T.; Chen, T.Q.; Zeng, Z.C.; Pan, Q.; Huang, W.; Han, C.; Fang, K.; Sun, L.Y.; Yang, Q.Q.; Wang, D.; et al. The lncRNA LAMP5-AS1 drives leukemia cell stemness by directly modulating DOT1L methyltransferase activity in MLL leukemia. J. Hematol. Oncol. 2020, 13, 78. [Google Scholar] [CrossRef]
  101. Orlandella, F.M.; Smaldone, G.; Salvatore, G.; Vitagliano, L.; Cianflone, A.; Parasole, R.; Beneduce, G.; Menna, G.; Salvatore, M.; Mirabelli, P. The lncRNA TEX41 is upregulated in pediatric B-Cells Acute Lymphoblastic Leukemia and it is necessary for leukemic cell growth. Biomark. Res. 2021, 9, 54. [Google Scholar] [CrossRef]
  102. Fernando, T.R.; Rodriguez-Malave, N.I.; Waters, E.V.; Yan, W.; Casero, D.; Basso, G.; Pigazzi, M.; Rao, D.S. LncRNA Expression Discriminates Karyotype and Predicts Survival in B-Lymphoblastic Leukemia. Mol. Cancer Res. 2015, 13, 839–851. [Google Scholar] [CrossRef] [Green Version]
  103. Zhang, L.; Zhou, S.; Zhou, T.; Li, X.; Tang, J. Targeting the lncRNA DUXAP8/miR-29a/PIK3CA Network Restores Doxorubicin Chemosensitivity via PI3K-AKT-mTOR Signaling and Synergizes With Inotuzumab Ozogamicin in Chemotherapy-Resistant B-Cell Acute Lymphoblastic Leukemia. Front. Oncol. 2022, 12, 773601. [Google Scholar] [CrossRef]
  104. Zhao, D.; Xing, Q.; Song, H.; Zhao, Y.; Guo, G. LINC00265/miR-4500 Axis Accelerates Acute Lymphoblastic Leukemia Progression by Enhancing STAT3 Signals. Cancer Manag. Res. 2021, 13, 8147–8156. [Google Scholar] [CrossRef]
  105. Li, G.; Lei, X.; Zhang, Y.; Liu, Z.; Zhu, K. LncRNA PPM1A-AS Regulate Tumor Development Through Multiple Signal Pathways in T-Cell Acute Lymphoblastic Leukemia. Front. Oncol. 2021, 11, 761205. [Google Scholar] [CrossRef]
  106. Zeng, P.; Chai, Y.; You, C.; Yue, L.; Wu, C.; Chen, H.; Li, L.; Li, J.; Liu, H.; Zhang, Y.; et al. Correlation analysis of long non-coding RNA TUG1 with disease risk, clinical characteristics, treatment response, and survival profiles of adult Ph Acute lymphoblastic leukemia. J. Clin. Lab. Anal. 2021, 35, e23583. [Google Scholar] [CrossRef]
  107. Abuduer, M.; A, E.Z.G. LINC00665 promotes the viability, migration and invasion of T cell acute lymphoblastic leukemia cells by targeting miR-101 via modulating PI3K/Akt pathway. Tissue Cell 2021, 71, 101579. [Google Scholar] [CrossRef]
  108. Mao, J.; Gao, W.; Xue, L.; Wang, J.; Zhao, L. The lncRNA SLCO4A1-AS1/miR-876-3p/RBBP6 axis regulates cell proliferation and apoptosis in acute lymphocytic leukemia via the JNK signaling pathway. Int. J. Lab. Hematol. 2021, 43, 1050–1061. [Google Scholar] [CrossRef]
  109. Ju, J.K.; Han, W.N.; Shi, C.L. Long non-coding RNA (lncRNA) plasmacytoma variant translocation 1 gene (PVT1) modulates the proliferation and apoptosis of acute lymphoblastic leukemia cells by sponging miR-486-5p. Bioengineered 2022, 13, 4587–4597. [Google Scholar] [CrossRef]
  110. Yazdi, N.; Houshmand, M.; Atashi, A.; Kazemi, A.; Najmedini, A.A.; Zarif, M.N. Long noncoding RNA PVT1: Potential oncogene in the development of acute lymphoblastic leukemia. Turk. J. Biol. 2018, 42, 405–413. [Google Scholar] [CrossRef]
  111. Yang, J.; Yang, Y. Long noncoding RNA endogenous bornavirus-like nucleoprotein acts as an oncogene by regulating microRNA-655-3p expression in T-cell acute lymphoblastic leukemia. Bioengineered 2022, 13, 6409–6419. [Google Scholar] [CrossRef]
  112. Garitano-Trojaola, A.; José-Enériz, E.S.; Ezponda, T.; Unfried, J.P.; Carrasco-León, A.; Razquin, N.; Barriocanal, M.; Vilas-Zornoza, A.; Sangro, B.; Segura, V.; et al. Deregulation of linc-PINT in acute lymphoblastic leukemia is implicated in abnormal proliferation of leukemic cells. Oncotarget 2018, 9, 12842–12852. [Google Scholar] [CrossRef]
  113. Zhang, Y.Y.; Huang, S.H.; Zhou, H.R.; Chen, C.J.; Tian, L.H.; Shen, J.Z. Role of HOTAIR in the diagnosis and prognosis of acute leukemia. Oncol. Rep. 2016, 36, 3113–3122. [Google Scholar] [CrossRef] [Green Version]
  114. Zhao, Q.; Zhao, S.; Li, J.; Zhang, H.; Qian, C.; Wang, H.; Liu, J.; Zhao, Y. TCF7L2 activated HOXA-AS2 decreased the glucocorticoid sensitivity in acute lymphoblastic leukemia through regulating HOXA3/EGFR/Ras/Raf/ MEK/ERK pathway. Biomed. Pharmacother. 2019, 109, 1640–1649. [Google Scholar] [CrossRef]
  115. Yang, T.; Jin, X.; Lan, J.; Wang, W. Long non-coding RNA SNHG16 has Tumor suppressing effect in acute lymphoblastic leukemia by inverse interaction on hsa-miR-124-3p. IUBMB Life 2019, 71, 134–142. [Google Scholar] [CrossRef] [Green Version]
  116. Song, Y.; Guo, N.H.; Zheng, J.F. LncRNA-MALAT1 regulates proliferation and apoptosis of acute lymphoblastic leukemia cells via miR-205-PTK7 pathway. Pathol. Int. 2020, 70, 724–732. [Google Scholar] [CrossRef]
  117. Melo, C.P.; Campos, C.B.; Rodrigues, J.; Aguirre-Neto, J.C.; Atalla, Â.; Pianovski, M.A.; Carbone, E.K.; Lares, L.B.; Moraes-Souza, H.; Octacílio-Silva, S.; et al. Long non-coding RNAs: Bioarkers for acute leukaemia subtypes. Br. J. Haematol. 2016, 173, 318–320. [Google Scholar] [CrossRef]
  118. Wallaert, A.; Durinck, K.; Van Loocke, W.; Van de Walle, I.; Matthijssens, F.; Volders, P.J.; Avila Cobos, F.; Rombaut, D.; Rondou, P.; Mestdagh, P.; et al. Long noncoding RNA signatures define oncogenic subtypes in T-cell acute lymphoblastic leukemia. Leukemia 2016, 30, 1927–1930. [Google Scholar] [CrossRef]
  119. Arthur, G.; Almamun, M.; Taylor, K. Hypermethylation of antisense long noncoding RNAs in acute lymphoblastic leukemia. Epigenomics 2017, 9, 635–645. [Google Scholar] [CrossRef]
  120. Cheng, H.; Huang, C.M.; Wang, Y.; Hu, X.X.; Xu, X.Q.; Song, X.M.; Tang, G.S.; Chen, L.; Yang, J.M. Microarray profiling and co-expression network analysis of the lncRNAs and mRNAs associated with acute leukemia in adults. Mol. Biosyst. 2017, 13, 1102–1108. [Google Scholar] [CrossRef]
  121. Lajoie, M.; Drouin, S.; Caron, M.; St-Onge, P.; Ouimet, M.; Gioia, R.; Lafond, M.H.; Vidal, R.; Richer, C.; Oualkacha, K.; et al. Specific expression of novel long non-coding RNAs in high-hyperdiploid childhood acute lymphoblastic leukemia. PLoS ONE 2017, 12, e0174124. [Google Scholar] [CrossRef] [Green Version]
  122. Caron, M.; St-Onge, P.; Drouin, S.; Richer, C.; Sontag, T.; Busche, S.; Bourque, G.; Pastinen, T.; Sinnett, D. Very long intergenic non-coding RNA transcripts and expression profiles are associated to specific childhood acute lymphoblastic leukemia subtypes. PLoS ONE 2018, 13, e0207250. [Google Scholar] [CrossRef]
  123. Cuadros, M.; Andrades, Á.; Coira, I.F.; Baliñas, C.; Rodríguez, M.I.; Álvarez-Pérez, J.C.; Peinado, P.; Arenas, A.M.; García, D.J.; Jiménez, P.; et al. Expression of the long non-coding RNA TCL6 is associated with clinical outcome in pediatric B-cell acute lymphoblastic leukemia. Blood Cancer J. 2019, 9, 93. [Google Scholar] [CrossRef] [Green Version]
  124. James, A.R.; Schroeder, M.P.; Neumann, M.; Bastian, L.; Eckert, C.; Gökbuget, N.; Tanchez, J.O.; Schlee, C.; Isaakidis, K.; Schwartz, S.; et al. Long non-coding RNAs defining major subtypes of B cell precursor acute lymphoblastic leukemia. J. Hematol. Oncol. 2019, 12, 8. [Google Scholar] [CrossRef] [Green Version]
  125. Li, S.; Bian, H.; Cao, Y.; Juan, C.; Cao, Q.; Zhou, G.; Fang, Y. Identification of novel lncRNAs involved in the pathogenesis of childhood acute lymphoblastic leukemia. Oncol. Lett. 2019, 17, 2081–2090. [Google Scholar] [CrossRef] [Green Version]
  126. Affinito, O.; Pane, K.; Smaldone, G.; Orlandella, F.M.; Mirabelli, P.; Beneduce, G.; Parasole, R.; Ripaldi, M.; Salvatore, M.; Franzese, M. lncRNAs-mRNAs Co-Expression Network Underlying Childhood B-Cell Acute Lymphoblastic Leukaemia: A Pilot Study. Cancers 2020, 12, 2489. [Google Scholar] [CrossRef]
  127. Bárcenas-López, D.A.; Núñez-Enríquez, J.C.; Hidalgo-Miranda, A.; Beltrán-Anaya, F.O.; May-Hau, D.I.; Jiménez-Hernández, E.; Bekker-Méndez, V.C.; Flores-Lujano, J.; Medina-Sansón, A.; Tamez-Gómez, E.L.; et al. Transcriptome Analysis Identifies LINC00152 as a Biomarker of Early Relapse and Mortality in Acute Lymphoblastic Leukemia. Genes 2020, 11, 302. [Google Scholar] [CrossRef] [Green Version]
  128. Cuadros, M.; García, D.J.; Andrades, A.; Arenas, A.M.; Coira, I.F.; Baliñas-Gavira, C.; Peinado, P.; Rodríguez, M.I.; Álvarez-Pérez, J.C.; Ruiz-Cabello, F.; et al. LncRNA-mRNA Co-Expression Analysis Identifies AL133346.1/CCN2 as Biomarkers in Pediatric B-Cell Acute Lymphoblastic Leukemia. Cancers 2020, 12, 3803. [Google Scholar] [CrossRef]
  129. Yu, W.; Wang, W.; Yu, X. Investigation of lncRNA-mRNA co-expression network in ETV6-RUNX1-positive pediatric B-cell acute lymphoblastic leukemia. PLoS ONE 2021, 16, e0253012. [Google Scholar] [CrossRef]
  130. Das Chagas, P.F.; de Sousa, G.R.; Kodama, M.H.; de Biagi Junior, C.; Yunes, J.A.; Brandalise, S.R.; Calin, G.A.; Tone, L.G.; Scrideli, C.A.; de Oliveira, J.C. Ultraconserved long non-coding RNA uc.112 is highly expressed in childhood T versus B-cell acute lymphoblastic leukemia. Hematol. Transfus. Cell Ther. 2021, 43, 28–34. [Google Scholar] [CrossRef]
  131. Wang, W.; Lyu, C.; Wang, F.; Wang, C.; Wu, F.; Li, X.; Gan, S. Identification of Potential Signatures and Their Functions for Acute Lymphoblastic Leukemia: A Study Based on the Cancer Genome Atlas. Front. Genet. 2021, 12, 656042. [Google Scholar] [CrossRef] [PubMed]
  132. Xia, J.; Wang, M.; Zhu, Y.; Bu, C.; Li, T. Differential mRNA and long noncoding RNA expression profiles in pediatric B-cell acute lymphoblastic leukemia patients. BMC Pediatr. 2022, 22, 10. [Google Scholar] [CrossRef] [PubMed]
  133. Ngoc, P.C.T.; Tan, S.H.; Tan, T.K.; Chan, M.M.; Li, Z.; Yeoh, A.; Tenen, D.G.; Sanda, T. Identification of novel lncRNAs regulated by the TAL1 complex in T-cell acute lymphoblastic leukemia. Leukemia 2018, 32, 2138–2151. [Google Scholar] [CrossRef] [PubMed]
  134. Verboom, K.; Van Loocke, W.; Volders, P.J.; Decaesteker, B.; Cobos, F.A.; Bornschein, S.; de Bock, C.E.; Atak, Z.K.; Clappier, E.; Aerts, S.; et al. A comprehensive inventory of TLX1 controlled long non-coding RNAs in T-cell acute lymphoblastic leukemia through polyA+ and total RNA sequencing. Haematologica 2018, 103, e585–e589. [Google Scholar] [CrossRef]
  135. Sanda, T.; Lawton, L.N.; Barrasa, M.I.; Fan, Z.P.; Kohlhammer, H.; Gutierrez, A.; Ma, W.; Tatarek, J.; Ahn, Y.; Kelliher, M.A.; et al. Core transcriptional regulatory circuit controlled by the TAL1 complex in human T cell acute lymphoblastic leukemia. Cancer Cell 2012, 22, 209–221. [Google Scholar] [CrossRef] [Green Version]
  136. Grabher, C.; von Boehmer, H.; Look, A.T. Notch 1 activation in the molecular pathogenesis of T-cell acute lymphoblastic leukaemia. Nat. Rev. Cancer 2006, 6, 347–359. [Google Scholar] [CrossRef]
  137. Durinck, K.; Wallaert, A.; Van de Walle, I.; Van Loocke, W.; Volders, P.J.; Vanhauwaert, S.; Geerdens, E.; Benoit, Y.; Van Roy, N.; Poppe, B.; et al. The Notch driven long non-coding RNA repertoire in T-cell acute lymphoblastic leukemia. Haematologica 2014, 99, 1808–1816. [Google Scholar] [CrossRef] [Green Version]
  138. Renneville, A.; Gasser, J.A.; Grinshpun, D.E.; Jean Beltran, P.M.; Udeshi, N.D.; Matyskiela, M.E.; Clayton, T.; McConkey, M.; Viswanathan, K.; Tepper, A.; et al. Avadomide induces degradation of ZMYM2 fusion oncoproteins in hematologic malignancies. Blood Cancer Discov. 2021, 2, 250–265. [Google Scholar] [CrossRef]
  139. Tsuzuki, S.; Yasuda, T.; Kojima, S.; Kawazu, M.; Akahane, K.; Inukai, T.; Imaizumi, M.; Morishita, T.; Miyamura, K.; Ueno, T.; et al. Targeting MEF2D-fusion Oncogenic Transcriptional Circuitries in B-cell Precursor Acute Lymphoblastic Leukemia. Blood Cancer Discov. 2020, 1, 82–95. [Google Scholar] [CrossRef]
  140. Montaño, A.; Ordoñez, J.L.; Alonso-Pérez, V.; Hernández-Sánchez, J.; Santos, S.; González, T.; Benito, R.; García-Tuñón, I.; Hernández-Rivas, J.M. ETV6/RUNX1 Fusion Gene Abrogation Decreases the Oncogenicity of Tumour Cells in a Preclinical Model of Acute Lymphoblastic Leukaemia. Cells 2020, 9, 215. [Google Scholar] [CrossRef] [Green Version]
  141. Ghazavi, F.; De Moerloose, B.; Van Loocke, W.; Wallaert, A.; Helsmoortel, H.H.; Ferster, A.; Bakkus, M.; Plat, G.; Delabesse, E.; Uyttebroeck, A.; et al. Unique long non-coding RNA expression signature in ETV6/RUNX1-driven B-cell precursor acute lymphoblastic leukemia. Oncotarget 2016, 7, 73769–73780. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. El Chaer, F.; Keng, M.; Ballen, K.K. MLL-Rearranged Acute Lymphoblastic Leukemia. Curr. Hematol. Malig. Rep. 2020, 15, 83–89. [Google Scholar] [CrossRef] [PubMed]
  143. Fang, K.; Han, B.W.; Chen, Z.H.; Lin, K.Y.; Zeng, C.W.; Li, X.J.; Li, J.H.; Luo, X.Q.; Chen, Y.Q. A distinct set of long non-coding RNAs in childhood MLL-rearranged acute lymphoblastic leukemia: Biology and epigenetic target. Hum. Mol. Genet. 2014, 23, 3278–3288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Becker, J.C.; Andersen, M.H.; Schrama, D.; Thor Straten, P. Immune-suppressive properties of the tumor microenvironment. Cancer Immunol. Immunother. 2013, 62, 1137–1148. [Google Scholar] [CrossRef] [PubMed]
  145. Niesel, K.; Schulz, M.; Anthes, J.; Alekseeva, T.; Macas, J.; Salamero-Boix, A.; Möckl, A.; Oberwahrenbrock, T.; Lolies, M.; Stein, S.; et al. The immune suppressive microenvironment affects efficacy of radio-immunotherapy in brain metastasis. EMBO Mol. Med. 2021, 13, e13412. [Google Scholar] [CrossRef]
  146. Haloupek, N. The Landscape of Blood Cancer Research Today—And Where the Field Is Headed. Blood Cancer Discov. 2020, 1, 1–4. [Google Scholar] [CrossRef]
  147. Wang, Y.; Yang, X.; Sun, X.; Rong, L.; Kang, M.; Wu, P.; Ji, X.; Lin, R.; Huang, J.; Xue, Y.; et al. Bone marrow infiltrated Lnc-INSR induced suppressive immune microenvironment in pediatric acute lymphoblastic leukemia. Cell Death Dis. 2018, 9, 1043. [Google Scholar] [CrossRef]
  148. Zhang, Q.; Hu, H.; Chen, S.Y.; Liu, C.J.; Hu, F.F.; Yu, J.; Wu, Y.; Guo, A.Y. Transcriptome and Regulatory Network Analyses of CD19-CAR-T Immunotherapy for B-ALL. Genom. Proteom. Bioinform. 2019, 17, 190–200. [Google Scholar] [CrossRef]
  149. Hashemi, M.; Bahari, G.; Naderi, M.; Sadeghi Bojd, S.; Taheri, M. Association of lnc-LAMC2-1:1 rs2147578 and CASC8 rs10505477 Polymorphisms with Risk of Childhood Acute Lymphoblastic Leukemia. Asian Pac. J. Cancer Prev. 2016, 17, 4985–4989. [Google Scholar]
  150. Han, J.; Zhou, W.; Jia, M.; Wen, J.; Jiang, J.; Shi, J.; Zhang, K.; Ma, H.; Liu, J.; Ren, J.; et al. Expression quantitative trait loci in long non-coding RNA PAX8-AS1 are associated with decreased risk of cervical cancer. Mol. Genet. Genom. 2016, 291, 1743–1748. [Google Scholar] [CrossRef]
  151. Bahari, G.; Hashemi, M.; Naderi, M.; Sadeghi-Bojd, S.; Taheri, M. Long non-coding RNA PAX8-AS1 polymorphisms increase the risk of childhood acute lymphoblastic leukemia. Biomed. Rep. 2018, 8, 184–190. [Google Scholar] [CrossRef] [PubMed]
  152. Pei, J.S.; Chang, W.S.; Chen, C.C.; Mong, M.C.; Hsu, S.W.; Hsu, P.C.; Hsu, Y.N.; Wang, Y.C.; Tsai, C.W.; Bau, D.T. Novel Contribution of Long Non-coding RNA MEG3 Genotype to Prediction of Childhood Leukemia Risk. Cancer Genom. Proteom. 2022, 19, 27–34. [Google Scholar] [CrossRef] [PubMed]
  153. Gasic, V.; Stankovic, B.; Zukic, B.; Janic, D.; Dokmanovic, L.; Krstovski, N.; Lazic, J.; Milosevic, G.; Lucafò, M.; Stocco, G.; et al. Expression Pattern of Long Non-coding RNA Growth Arrest-specific 5 in the Remission Induction Therapy in Childhood Acute Lymphoblastic Leukemia. J. Med. Biochem. 2019, 38, 292–298. [Google Scholar] [CrossRef] [PubMed]
  154. Gioia, R.; Drouin, S.; Ouimet, M.; Caron, M.; St-Onge, P.; Richer, C.; Sinnett, D. LncRNAs downregulated in childhood acute lymphoblastic leukemia modulate apoptosis, cell migration, and DNA damage response. Oncotarget 2017, 8, 80645–80650. [Google Scholar] [CrossRef] [Green Version]
  155. Qi, H.; Chi, L.; Wang, X.; Jin, X.; Wang, W.; Lan, J. Identification of a Seven-lncRNA-mRNA Signature for Recurrence and Prognostic Prediction in Relapsed Acute Lymphoblastic Leukemia Based on WGCNA and LASSO Analyses. Anal. Cell. Pathol. 2021, 2021, 6692022. [Google Scholar] [CrossRef]
  156. Pouyanrad, S.; Rahgozar, S.; Ghodousi, E.S. Dysregulation of miR-335-3p, targeted by NEAT1 and MALAT1 long non-coding RNAs, is associated with poor prognosis in childhood acute lymphoblastic leukemia. Gene 2019, 692, 35–43. [Google Scholar] [CrossRef]
  157. Chen, Z.; Yang, F.; Liu, H.; Fan, F.; Lin, Y.; Zhou, J.; Cai, Y.; Zhang, X.; Wu, Y.; Mao, R.; et al. Identification of a nomogram based on an 8-lncRNA signature as a novel diagnostic biomarker for childhood acute lymphoblastic leukemia. Aging 2021, 13, 15548–15568. [Google Scholar] [CrossRef]
  158. Gao, W. Long non-coding RNA MEG3 as a candidate prognostic factor for induction therapy response and survival profile in childhood acute lymphoblastic leukemia patients. Scand. J. Clin. Lab. Investig. 2021, 81, 194–200. [Google Scholar] [CrossRef]
  159. Xagorari, M.; Marmarinos, A.; Kossiva, L.; Baka, M.; Doganis, D.; Servitzoglou, M.; Tsolia, M.; Scorilas, A.; Avgeris, M.; Gourgiotis, D. Overexpression of the GR Riborepressor LncRNA GAS5 Results in Poor Treatment Response and Early Relapse in Childhood B-ALL. Cancers 2021, 13, 6064. [Google Scholar] [CrossRef]
  160. Memczak, S.; Jens, M.; Elefsinioti, A.; Torti, F.; Krueger, J.; Rybak, A.; Maier, L.; Mackowiak, S.D.; Gregersen, L.H.; Munschauer, M.; et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature 2013, 495, 333–338. [Google Scholar] [CrossRef]
  161. Verduci, L.; Tarcitano, E.; Strano, S.; Yarden, Y.; Blandino, G. CircRNAs: Role in human diseases and potential use as biomarkers. Cell Death Dis. 2021, 12, 468. [Google Scholar] [CrossRef] [PubMed]
  162. Zhang, X.O.; Wang, H.B.; Zhang, Y.; Lu, X.; Chen, L.L.; Yang, L. Complementary sequence-mediated exon circularization. Cell 2014, 159, 134–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Starke, S.; Jost, I.; Rossbach, O.; Schneider, T.; Schreiner, S.; Hung, L.H.; Bindereif, A. Exon circularization requires canonical splice signals. Cell Rep. 2015, 10, 103–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Zhang, Y.; Zhang, X.O.; Chen, T.; Xiang, J.F.; Yin, Q.F.; Xing, Y.H.; Zhu, S.; Yang, L.; Chen, L.L. Circular intronic long noncoding RNAs. Mol. Cell 2013, 51, 792–806. [Google Scholar] [CrossRef] [Green Version]
  165. Zhang, Y.; Xue, W.; Li, X.; Zhang, J.; Chen, S.; Zhang, J.L.; Yang, L.; Chen, L.L. The Biogenesis of Nascent Circular RNAs. Cell Rep. 2016, 15, 611–624. [Google Scholar] [CrossRef] [Green Version]
  166. Van Zonneveld, A.J.; Kölling, M.; Bijkerk, R.; Lorenzen, J.M. Circular RNAs in kidney disease and cancer. Nat. Rev. Nephrol. 2021, 17, 814–826. [Google Scholar] [CrossRef]
  167. Yang, Y.; Fan, X.; Mao, M.; Song, X.; Wu, P.; Zhang, Y.; Jin, Y.; Yang, Y.; Chen, L.L.; Wang, Y.; et al. Extensive translation of circular RNAs driven by N6-methyladenosine. Cell Res. 2017, 27, 626–641. [Google Scholar] [CrossRef] [Green Version]
  168. Rybak-Wolf, A.; Stottmeister, C.; Glažar, P.; Jens, M.; Pino, N.; Giusti, S.; Hanan, M.; Behm, M.; Bartok, O.; Ashwal-Fluss, R.; et al. Circular RNAs in the Mammalian Brain Are Highly Abundant, Conserved, and Dynamically Expressed. Mol. Cell 2015, 58, 870–885. [Google Scholar] [CrossRef] [Green Version]
  169. Huang, L.; Rong, Y.; Tang, X.; Yi, K.; Wu, J.; Wang, F. Circular RNAs Are Promising Biomarkers in Liquid Biopsy for the Diagnosis of Non-small Cell Lung Cancer. Front. Mol. Biosci. 2021, 8, 625722. [Google Scholar] [CrossRef]
  170. Nicolet, B.P.; Engels, S.; Aglialoro, F.; van den Akker, E.; von Lindern, M.; Wolkers, M.C. Circular RNA expression in human hematopoietic cells is widespread and cell-type specific. Nucleic Acids Res. 2018, 46, 8168–8180. [Google Scholar] [CrossRef]
  171. Yu, C.Y.; Li, T.C.; Wu, Y.Y.; Yeh, C.H.; Chiang, W.; Chuang, C.Y.; Kuo, H.C. The circular RNA circBIRC6 participates in the molecular circuitry controlling human pluripotency. Nat. Commun. 2017, 8, 1149. [Google Scholar] [CrossRef] [PubMed]
  172. Li, J.; Sun, D.; Pu, W.; Wang, J.; Peng, Y. Circular RNAs in Cancer: Biogenesis, Function, and Clinical Significance. Trends Cancer 2020, 6, 319–336. [Google Scholar] [CrossRef] [PubMed]
  173. Jeha, S.; Choi, J.; Roberts, K.G.; Pei, D.; Coustan-Smith, E.; Inaba, H.; Rubnitz, J.E.; Ribeiro, R.C.; Gruber, T.A.; Raimondi, S.C.; et al. Clinical significance of novel subtypes of acute lymphoblastic leukemia in the context of minimal residual disease-directed therapy. Blood Cancer Discov. 2021, 2, 326–337. [Google Scholar] [CrossRef] [PubMed]
  174. Jiang, A.; Hilton, L.K.; Tang, J.; Rushton, C.K.; Grande, B.M.; Scott, D.W.; Morin, R.D. PRPS-ST: A protocol-agnostic self-training method for gene expression-based classification of blood cancers. Blood Cancer Discov. 2020, 1, 244–257. [Google Scholar] [CrossRef]
  175. Gaffo, E.; Boldrin, E.; Dal Molin, A.; Bresolin, S.; Bonizzato, A.; Trentin, L.; Frasson, C.; Debatin, K.M.; Meyer, L.H.; Te Kronnie, G.; et al. Circular RNA differential expression in blood cell populations and exploration of circRNA deregulation in pediatric acute lymphoblastic leukemia. Sci. Rep. 2019, 9, 14670. [Google Scholar] [CrossRef] [Green Version]
  176. Buratin, A.; Paganin, M.; Gaffo, E.; Dal Molin, A.; Roels, J.; Germano, G.; Siddi, M.T.; Serafin, V.; De Decker, M.; Gachet, S.; et al. Large-scale circular RNA deregulation in T-ALL: Unlocking unique ectopic expression of molecular subtypes. Blood Adv. 2020, 4, 5902–5914. [Google Scholar] [CrossRef]
  177. Guo, S.; Li, B.; Chen, Y.; Zou, D.; Yang, S.; Zhang, Y.; Wu, N.; Sheng, L.; Huang, H.; Ouyang, G.; et al. Hsa_circ_0012152 and Hsa_circ_0001857 Accurately Discriminate Acute Lymphoblastic Leukemia From Acute Myeloid Leukemia. Front. Oncol. 2020, 10, 1655. [Google Scholar] [CrossRef]
  178. Panda, A.C.; Grammatikakis, I.; Kim, K.M.; De, S.; Martindale, J.L.; Munk, R.; Yang, X.; Abdelmohsen, K.; Gorospe, M. Identification of senescence-associated circular RNAs (SAC-RNAs) reveals senescence suppressor CircPVT1. Nucleic Acids Res. 2017, 45, 4021–4035. [Google Scholar] [CrossRef]
  179. Chen, J.; Li, Y.; Zheng, Q.; Bao, C.; He, J.; Chen, B.; Lyu, D.; Zheng, B.; Xu, Y.; Long, Z.; et al. Circular RNA profile identifies circPVT1 as a proliferative factor and prognostic marker in gastric cancer. Cancer Lett. 2017, 388, 208–219. [Google Scholar] [CrossRef]
  180. Hu, J.; Han, Q.; Gu, Y.; Ma, J.; McGrath, M.; Qiao, F.; Chen, B.; Song, C.; Ge, Z. Circular RNA PVT1 expression and its roles in acute lymphoblastic leukemia. Epigenomics 2018, 10, 723–732. [Google Scholar] [CrossRef]
  181. Jia, Y.; Hu, H.; Gu, W. Regulation Effects of Circular RNA CircPVT1 and miR-125b on NF-κB Signal Pathway in Childhood ALL. Clin. Lab. 2021, 67. [Google Scholar] [CrossRef] [PubMed]
  182. Jia, Y.; Gu, W. Up-regulation of circPVT1 in T cell acute lymphoblastic leukemia promoted cell proliferation via miR-30e/DLL4 induced activating NOTCH signaling. Pathol. Res. Pract. 2021, 224, 153536. [Google Scholar] [CrossRef] [PubMed]
  183. Liu, X.; Zhou, C.; Li, Y.; Deng, Y.; Lu, W.; Li, J. Upregulation of circ-0000745 in acute lymphoblastic leukemia enhanced cell proliferation by activating ERK pathway. Gene 2020, 751, 144726. [Google Scholar] [CrossRef]
  184. Feng, H.; Li, F.; Tang, P. Circ_0000745 regulates NOTCH1-mediated cell proliferation and apoptosis in pediatric T-cell acute lymphoblastic leukemia through adsorbing miR-193b-3p. Hematology 2021, 26, 885–895. [Google Scholar] [CrossRef] [PubMed]
  185. Yang, X.; Li, Y.; Zhang, Y.; Liu, J. Circ_0000745 promotes acute lymphoblastic leukemia progression through mediating miR-494-3p/NET1 axis. Hematology 2022, 27, 11–22. [Google Scholar] [CrossRef]
  186. Zheng, Y.; Niu, B.; Zhang, W.; Ru, X.; Gao, Y.; Li, C.; Wu, X. Circular RNA circPRKCI contributes to malignant progression of T-cell acute lymphoblastic leukemia by modulating miR-20a-5p/SOX4 axis. Aging 2021, 13, 23757–23768. [Google Scholar] [CrossRef]
  187. Ling, Z.; Fang, Z.G.; Wu, J.Y.; Liu, J.J. The depletion of Circ-PRKDC enhances autophagy and apoptosis in T-cell acute lymphoblastic leukemia via microRNA-653-5p/Reelin mediation of the PI3K/AKT/mTOR signaling pathway. Kaohsiung J. Med. Sci. 2021, 37, 392–401. [Google Scholar] [CrossRef]
  188. Hou, Y.; Sun, J.; Huang, J.; Yao, F.; Chen, X.; Zhu, B.; Zhao, D. Circular RNA circRNA_0000094 sponges microRNA-223-3p and up-regulate F-box and WD repeat domain containing 7 to restrain T cell acute lymphoblastic leukemia progression. Hum Cell. 2021, 34, 977–989. [Google Scholar] [CrossRef]
  189. Tong, J.; Liu, H.; Yao, W.; Zhang, L.; Song, K.; Sun, G.; Sun, Z. Down-regulation of circRNA_0000143 regulates miR-142-3p/GRα axis to facilitate the progression of T-ALL. Cancer Sci. 2021. Epub ahead of print. [Google Scholar] [CrossRef]
  190. Zhu, Y.; Ma, X.; Zhang, H.; Wu, Y.; Kang, M.; Fang, Y.; Xue, Y. Mechanism of circADD2 as ceRNA in Childhood Acute Lymphoblastic Leukemia. Front. Cell Dev. Biol. 2021, 9, 639910. [Google Scholar] [CrossRef]
  191. He, A.T.; Liu, J.; Li, F.; Yang, B.B. Targeting circular RNAs as a therapeutic approach: Current strategies and challenges. Signal Transduct. Target. Ther. 2021, 6, 185. [Google Scholar] [CrossRef] [PubMed]
  192. Roels, J.; Thénoz, M.; Szarzyńska, B.; Landfors, M.; De Coninck, S.; Demoen, L.; Provez, L.; Kuchmiy, A.; Strubbe, S.; Reunes, L.; et al. Aging of preleukemic thymocytes drives CpG island hypermethylation in T-cell acute lymphoblastic leukemia. Blood Cancer Discov. 2020, 1, 274–289. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of major non-coding RNA types produced in cells. Size of each ncRNA type and main functions are included in the same panel as well. The figure is created with Biorender (Biorender.com (accessed on 28 January 2022)).
Figure 1. Schematic representation of major non-coding RNA types produced in cells. Size of each ncRNA type and main functions are included in the same panel as well. The figure is created with Biorender (Biorender.com (accessed on 28 January 2022)).
Ijms 23 04442 g001
Figure 2. Main biological functions of lncRNAs in cells. In general, lncRNAs with multiple exons are exported to the cytoplasm similar to mRNAs, while intronic lncRNAs are retained in the nucleus. Due to the flexible structure and long size, lncRNAs interact with DNA, RNA, and protein, thereby regulating chromatin state (histone modification, DNA methylation), transcription, pre-mRNA stability as well as splicing and processing, nuclear condensate formation (i.e., paraspeckles, nuclear speckles), mRNA stability, translation, and sponging miRNAs as well as orchestrating protein complex formation. The figure is created with Biorender (Biorender.com (accessed on 28 January 2022)).
Figure 2. Main biological functions of lncRNAs in cells. In general, lncRNAs with multiple exons are exported to the cytoplasm similar to mRNAs, while intronic lncRNAs are retained in the nucleus. Due to the flexible structure and long size, lncRNAs interact with DNA, RNA, and protein, thereby regulating chromatin state (histone modification, DNA methylation), transcription, pre-mRNA stability as well as splicing and processing, nuclear condensate formation (i.e., paraspeckles, nuclear speckles), mRNA stability, translation, and sponging miRNAs as well as orchestrating protein complex formation. The figure is created with Biorender (Biorender.com (accessed on 28 January 2022)).
Ijms 23 04442 g002
Figure 3. Main biogenesis pathways and functions of circRNAs in cells. (A), Biogenesis of exonic circRNA from back-splicing of pre-mRNA. RNA-binding proteins (RBPs) or orientation-opposite complementary sequences (OOCS) that form loop-like structures facilitate back-splicing and circRNAs formation; these circRNAs are generally exported to the cytoplasm and act as a miRNA sponge, protein complex coordinators as well as small peptide producers to modulate protein function; (B), Biogenesis of lncRNAs from intron lariats. A consensus RNA motif containing a 7-nt GU-rich element near the 5′ splice site and an 11-nt C-rich element near the branch point prevents detaching of the intron lariats and promotes intronic circRNA formation; these circRNAs are retained in the nucleus and mainly regulate Pol II-mediated transcription. The figure is created with Biorender (Biorender.com (accessed on 28 January 2022)).
Figure 3. Main biogenesis pathways and functions of circRNAs in cells. (A), Biogenesis of exonic circRNA from back-splicing of pre-mRNA. RNA-binding proteins (RBPs) or orientation-opposite complementary sequences (OOCS) that form loop-like structures facilitate back-splicing and circRNAs formation; these circRNAs are generally exported to the cytoplasm and act as a miRNA sponge, protein complex coordinators as well as small peptide producers to modulate protein function; (B), Biogenesis of lncRNAs from intron lariats. A consensus RNA motif containing a 7-nt GU-rich element near the 5′ splice site and an 11-nt C-rich element near the branch point prevents detaching of the intron lariats and promotes intronic circRNA formation; these circRNAs are retained in the nucleus and mainly regulate Pol II-mediated transcription. The figure is created with Biorender (Biorender.com (accessed on 28 January 2022)).
Ijms 23 04442 g003
Table 1. List of lncRNAs implicated in T-ALL pathogenesis and treatment.
Table 1. List of lncRNAs implicated in T-ALL pathogenesis and treatment.
lncRNASubjectFunctionAlterationTarget RegulationEffect on All Cell PhenotypeReferences
LUNAR1T-ALL cell lines and patient samplesoncogeneupregulatedenhancing IGF1R mRNA expression, sustaining IGF1 signalingpromoting cell growth via acting as NOTCH effector[77,78]
T-ALL-R-LncR1T-ALL cell lines and patient samplesoncogeneupregulatedinhibiting pro-apoptotic factor Par-4/THAP1 protein complexinhibiting apoptosis[79]
NALT1T-ALL cell lines and patient samplesoncogeneupregulatedcausing transcriptional activation of NOTCH1 signalingpromoting cell proliferation[80]
LINC00478T-ALL cell lines and patient samplesoncogeneupregulatedupregulating miR-125b productionpromoting cell growth and invasiveness[81]
ARIELT-ALL cell lines and patient samplesoncogeneupregulatedenhancing ARID5B gene expression promoting cell growth and survival[82]
CDKN2B-AS1T-ALL cell lines and patient samplesoncogeneupregulatedsponging miR-335-3p to upregulate TRAF5promoting cell proliferation and cell cycle progression[83]
LINC00511T-ALL cell lines and patient samplesoncogeneupregulated sponging miR-195-5p to upregulate LRRK1boosting cell proliferation and invasion, inhibiting apoptosis[84]
ANRILT-ALL cell lines and patient samplesoncogeneupregulatedsuppressing miR-7-5p to upregulate TCF4promoting cell viability, migration, and invasion [85]
AWPPHT-ALL cell lines and patient samplesoncogeneupregulatedupregulation of ROCK2 expressionpromoting proliferation and inhibiting apoptosis[86]
NEAT1T-ALL cell lines and patient samplesoncogeneupregulatedsponging miR-146b-5p to upregulate expression of NOTCH1promoting cell proliferation and growth[87]
H19T-ALL cell lines and patient samplesoncogeneupregulatedinducing SOX2, OCT-4, and NANOG expression; decreasing miR-326 level to upregulate BCL-2 expressionmaintaining stemness and promoting cell proliferation; inhibiting apoptosis[88,89,90]
LINC00853T-ALL cell lines and patient samplestumor suppressordownregulatedupregulating CCR9 expressioninhibiting cell proliferation, migration, and invasion[91]
LNC00221T-ALL cell lines Jurkat, CCRF-CEM, CEM/C1tumor suppressordownregulatedsponging miR-152-3p to upregulate ATPA2Ainhibiting cell proliferation and apoptosis[92]
VPS9D1-AS1T-ALL cell lines Molt-3 and Molt-4oncogeneupregulatedinhibiting miR-419-5p, miR-214-3p to upregulate GPX1inhibiting apoptosis and promoting survival[93]
Abbreviations: LUNAR1, leukemia-induced noncoding activator RNA-1; T-ALL-R-LncR1, T-ALL-related long non-coding RNA; NALT, NOTCH1-associated lncRNA in T-cell acute lymphoblastic leukemia; LINC00478, long intergenic non-protein coding RNA 478; ARIEL, ARID5B-inducing enhancer associated long noncoding RNA; CDKN2B-AS1, CDKN2B antisense RNA 1; LINC00511, long intergenic non-protein coding RNA 511; ANRIL, antisense non-coding RNA in the INK4 locus; AWPPH, associated with poor prognosis of hepatocellular carcinoma; NEAT1, nuclear paraspeckle assembly transcript 1; LINC00853, long intergenic non-protein coding RNA 583; LNC00221, long intergenic non-protein coding RNA 211; VPS9D1-AS1, VPS9D1 antisense RNA 1.
Table 2. List of lncRNAs implicated in B-ALL pathogenesis and treatment.
Table 2. List of lncRNAs implicated in B-ALL pathogenesis and treatment.
lncRNASubjectFunctionAlterationTarget RegulationEffect on ALL Cell PhenotypeReference
RP11-137H2.4B-ALL cell lines and patient samplesoncogeneupregulatedmodulated expression of NRAS/BRAF/NF-κB MAPK cascade and cell cycle pathwaysenhancing cell proliferation, migration, and apoptosis, inducing GC resistance[94]
CASC15RUNX1/AML+ B-ALL cell lines and patient samplesoncogeneupregulatedenhancing YY1-mediated regulation of the SOX4 promoterpromoting cell proliferation and survival[95]
ZEB1-AS1B-ALL cell lines and patient samplesoncogeneupregulatedbinding to IL-11 and promoting IL-11 stability, activating STAT3increasing cell proliferation[96]
BALR-6B-ALL cell lines and patient samplesoncogeneupregulatedenhancing SP1-mediated transcription of CREB1increasing proliferation and decreasing apoptosis[97]
IURPh+ B-ALL patient samples and cell linestumor suppressordownregulatedinhibiting STAT5-CD71 pathway suppression of BCR-ABL1-mediated tumorigenesis [98]
CRNDEB-ALL cell lines and patient samplesoncogeneupregulatedsequestering miR-345-5p to upregulate CREBpromoting cell proliferation and inhibiting apoptosis[99]
LAMP5-AS1MLL rearranged B-ALL patient samples and cell linesoncogeneupregulatedpromoting methyltransferase activity of DOT1L to facilitate H3K9me2/me3 and increase HOXA expressionincreasing colony formation and inhibiting differentiation[100]
TEX41B-ALL patient samples and cell linesoncogeneupregulateddecreasing CDK4, CDK6 and p27 level, increasing p21 and p53 levelpromoting cell growth and inhibiting cell cycle[101]
BALR-2B-ALL patient samples and cell linesoncogeneupregulateddecreasing JUN and BIM expressionincreasing cell growth and conferring resistance to prednisone treatment[102]
DUXAP8B-ALL patient samples and cell linesoncogeneupregulatedsequestering miR-29a to increase PIK3CA expressionboosting proliferation, inhibiting apoptosis, and conferring Dox resistance[103]
Abbreviations: PVT1, plasmacytoma variant translocation 1; CASC15, cancer susceptibility candidate 15; ZEB1-AS1, ZEB1 antisense RNA 1; BALR-6, B-ALL-associated long RNA-6; IUR, imatinib upregulated; CRNDE, colorectal neoplasia differentially expressed; LAMP5-AS1, LAMP5 antisense RNA1; TEX41, testis expressed 41; BALR-2, B-ALL-associated long RNA-2; DUXAP8, double homeobox A pseudogene 8.
Table 3. List of lncRNAs implicated in ALL (B- or T-subtype not differentiated) pathogenesis and treatment.
Table 3. List of lncRNAs implicated in ALL (B- or T-subtype not differentiated) pathogenesis and treatment.
lncRNASubjectFunctionAlterationTarget RegulationEffect on ALL Cell PhenotypeReferences
LINC00265ALL patient samples and cell linesoncogeneupregulatedsponging miR-4500 to enhance STAT3 expressionfacilitating cell growth, proliferation, and migration[104]
PPM1A-ASALL patient samples and cell linesoncogeneupregulatedincreasing phosphorylation of STAT3, Akt, and Notch4promoting cell proliferation and inhibiting apoptosis[105]
TUG1Ph- ALL patient samplesoncogeneupregulatedunknownpromoting CNS infiltration[106]
LINC00665ALL patient samples and cell linesoncogeneupregulatedsuppressing miR-101 to activate PI3K/Akt pathwaypromoting cell viability, migration, and invasion [107]
SLCO4A1-AS1ALL patient samples and cell linesoncogeneupregulatedsponging miR-876-3p to upregulate RBBP6 and activate JNKinhibited cell proliferation and promoted apoptosis[108]
PVT1ALL patient samples and cell linesoncogeneupregulatedsponging miR-486-5p to increase MAML3 expression; increasing NOP2 and c-Myc, decreasing p15, p16increasing cell viability restraining apoptosis; deregulating cell cycle, inhibiting apoptosis [109,110]
EBLN3PALL patient samples and cell linesoncogeneupregulatednegatively regulating miR-655-3p promoting cell proliferation, invasion, and migration[111]
PINTALL patient samples and cell linestumor suppressordownregulatedincreasing transcription of HMOX1inducing apoptosis and causing cell cycle arrest[112]
HOTAIRALL patient samplesoncogeneupregulatedincreasing EZH2, LSD1, DNMT3A and DNMT3B levelpromoting cell proliferation, survival, and migration[113]
HOXA-AS2ALL cell lines and patient samplesoncogeneupregulatedenhancing GC resistance via promoting HOXA3 expressionpromotion of cell proliferation, inhibition of apoptosis[114]
SNHG16ALL cell lines and patient samplesoncogeneupregulatedsponging miR-124-3ppromoting ALL cell proliferation and migration [115]
MALAT1ALL samples and cell linesoncogeneupregulatedsponging miR-205 to increase PTK7 expressionpromoting cell proliferation and apoptosis[116]
Abbreviations: LINC00265, long intergenic non-protein coding RNA 265; PPM1A-AS, protein phosphatase 1A antisense RNA; TUG1, Taurine upregulated gene 1; LINC00665, long intergenic non-protein coding RNA 665; SLCO4A1-AS1, SLCO4A1 antisense RNA; EBLN3P, endogenous Bornavirus-like nucleoprotein; PINT, p53-induced transcript; HOTAIR, HOX transcript antisense intergenic RNA; HOXA-AS2, HOXA cluster antisense RNA2; SNHG16, small nuclear RNA host gene 16; MALAT1, metastasis associated lung adenocarcinoma transcript 1.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Maimaitiyiming, Y.; Ye, L.; Yang, T.; Yu, W.; Naranmandura, H. Linear and Circular Long Non-Coding RNAs in Acute Lymphoblastic Leukemia: From Pathogenesis to Classification and Treatment. Int. J. Mol. Sci. 2022, 23, 4442. https://doi.org/10.3390/ijms23084442

AMA Style

Maimaitiyiming Y, Ye L, Yang T, Yu W, Naranmandura H. Linear and Circular Long Non-Coding RNAs in Acute Lymphoblastic Leukemia: From Pathogenesis to Classification and Treatment. International Journal of Molecular Sciences. 2022; 23(8):4442. https://doi.org/10.3390/ijms23084442

Chicago/Turabian Style

Maimaitiyiming, Yasen, Linyan Ye, Tao Yang, Wenjuan Yu, and Hua Naranmandura. 2022. "Linear and Circular Long Non-Coding RNAs in Acute Lymphoblastic Leukemia: From Pathogenesis to Classification and Treatment" International Journal of Molecular Sciences 23, no. 8: 4442. https://doi.org/10.3390/ijms23084442

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop