Abstract
Circular RNAs (circRNAs) are a new class of covalently closed RNA molecules whose 3’- and 5’-ends are linked by a back-splicing event. Emerging evidence has shown that circRNAs play a vital role in the occurrence and development of many diseases and are promising biomarkers and therapeutic targets. However, knowledge of circRNAs in hematological malignancies is limited. In this review, the biogenesis, categories, characteristics, and functions of circRNAs are summarized, especially the roles of circRNAs in hematopoiesis and hematological malignancies.
Keywords: circRNA, hematopoiesis, acute myeloid leukemia, chronic myeloid leukemia, lymphoid malignancies
Introduction
CircRNAs, first identified in 1976 in viruses to have a single-strand covalently closed structure, were considered viroids due to a lack of knowledge about them [1]. Subsequently, researchers discovered several circRNAs in different eukaryotic cells and speculated that they were generated by base-pairing between the two ends of RNA molecules [2]. In recent years, abundant evidence has shown that circRNAs are richly and widely expressed in eukaryotes and participate in most biological processes, including carcinogenesis. For example, serving as a sponge for miR-424-5p, circLARP4 suppresses cell proliferation and invasion of gastric cancer by regulating the expression of LAST1 [3]. CircCSNK1G3 can promote prostate cancer cell proliferation by acting as a stabilizer for miR-181b/d [4]. Ci-ANKRD52 has been suggested to regulate its parental gene by influencing the elongation of Pol II [5]. Some circRNAs, such as circZNF609 [6] and circ-MBL [7], function as templates for protein translation. A growing number of studies have shown that circRNAs are also involved in hematopoiesis and hematological malignancies. Although Bonizzato et al. [8] summarized the roles of circRNAs in hematopoiesis and hematological malignancies, they focused on the biogenesis and general functions of circRNAs due to the lack of research on hematological diseases at that time. Liu et al. [9] also summarized the roles of ncRNAs (noncoding RNAs) in acute myeloid leukemia, but they focused on acute myeloid leukemia and noncoding RNAs. This article provides a comprehensive outlook on circRNAs from their biological features such as biogenesis, categories, characteristics and functions to their roles in hematopoiesis and hematological malignancies. Although circRNAs have been shown to play a variety of roles in hematological diseases, our understanding of circRNAs may be just the tip of the iceberg.
Biogenesis and categories of circRNAs
The biogenesis of circRNAs can occur during and after transcription by a back-splicing process [10]. According to their different origins, four types of circRNAs have been found, namely, circRNA from pre-mRNA, tricRNA (tRNA intronic circRNA) [11-13] from pre-tRNA, f-circRNA (fusion-circRNA) [14] from gene fusions, and rt-circRNA [15,16] from transcription read-through. In addition, circRNAs originating from pre-mRNAs can be divided into four subcategories according to their composition: exonic circRNA (ecircRNA), circular intronic RNA (ciRNA), exon-intron circRNA (EIciRNA), and intergenic circular RNA (intergenic circRNA). The classification of circRNAs is shown in Table 1.
Table 1.
CircRNA | Origin | Components | Location | Potential function |
---|---|---|---|---|
ecircRNA | pre-mRNA | exon | cytoplasm | biomarker, miRNA sponge, translating, etc. |
ciRNA | pre-mRNA | intron | nucleus | regulating splicing and transcription |
EIciRNA | pre-mRNA | exon and intron | nucleus | regulating splicing and transcription |
intergenic circRNA | pre-mRNA | intergenic sequence | - | - |
tricRNA | intron-containing pre-tRNA | intron from pre-tRNA | - | - |
f-circRNA | fusion gene | two or more genes from different strands | - | oncogene |
rt-circRNA | read-through transcript | two or more adjacent gene from the same strand | - | - |
EcircRNAs and EIciRNAs may have the same biogenetic mechanisms [17]. Three mechanisms have been proposed for their biogenesis: direct back-splicing or intron pairing-driven circularization [18] (Figure 1A), exon-skipping or lariat-driven circularization [18] (Figure 1B), and RNA-binding protein-driven circularization [19] (Figure 1C). CiRNA biogenesis relies on a motif consisting of a 7 nt GU-rich component near the 5’ splicing site and an 11 nt AC-rich element near the branchpoint site (Figure 1D) [5]. An intergenic circRNA derived from chromosome 5 was detected by CIRI (CircRNA Identification, a new circRNA identification tool). It contains two intronic circRNA fragments, which are spliced at the flanking GT-AG splicing signals to form a complete circRNA (Figure 1E) [20]. The intron-containing pre-tRNAs are cleaved into exon and intron termini by specific motifs and enzymes, and then the intron termini can be circularized to form tricRNA [11,13] (Figure 1F). F-circRNAs are generated from translocated or rearranged chromosomes, which results in the connection of two otherwise isolated genes. Subsequently, the flanking intron complementary sequences of the junction point allow the formation of a f-circRNA (Figure 1G) [14]. Rt-circRNAs are a novel class of circRNAs that contain several exons stemming from different genes [15,16]. In some cases, such as the pre-mRNA 3’ processing was suppressed, RNA Pol II can extend to the next gene, which produces read-through transcripts containing two or more adjacent genes [15], after which rt-circRNA can be formed by back-splicing when the intronic repeats are present in the 5’ end of the upstream gene and the 3’ end of the downstream gene (Figure 1J) [15,16].
Characteristics of circRNAs
To date, circRNAs have been confirmed to have at least the following characteristics. (1) Stability: the circRNA closed-loop structure without a 5’ cap and 3’ tail grants them resistance to RNase R or RNA exonuclease [21]. CircRNAs have a half-life of approximately 24 h, while mRNAs have a half-life of approximately 4-6 h at 4°C [22]. (2) Specificity: circRNAs have cell-type-specific and tissue-specific expression patterns [23]. (3) Abundance: circRNAs are extremely abundant, some with circular to linear ratios greater than 1 [24]. (4) Conservation: circRNAs are evolutionarily conserved in different species [25]. (5) Distribution: circRNAs can be detected in multiple tissues, such as peripheral blood [19], bone marrow [26-30] and exosomes [31].
Functions of circRNAs
Functions of the vast majority of circRNAs remain unclear, but increasing studies have revealed that circRNAs play a crucial role in most biological processes by regulating miRNAs or alternative splicing patterns, interacting with RBPs (RNA-binding proteins), producing proteins, etc. The potential functions of circRNAs are shown in Figure 2.
Serving as miRNA sponges, stabilizers, or traps
CircRNAs can act as miRNA sponges to sequester miRNAs, stabilize miRNAs, or trap miRNAs for storage or transport, thereby regulating the expression of downstream genes. The most typical case is CDR1as (CDR1 antisense), which contains over 70 miR-7 binding sites and a number of almost perfectly complementary miR-671 binding sites. CDR1as can act as a sponge or stabilizer to bind miR-7, leading to the downregulation of miR-7 and thereby restoring the expression of miR-7 target genes [32,33]. Additionally, by binding to miR-671, CDR1as directs its own Argonaute 2 (AGO2)-mediated cleavage, thereby releasing its miR-7 cargo [33,34].
Interacting with RNA-binding proteins
CircRNAs interact with RBPs to form circRNPs, which store, sequester, translocate, and sort proteins and further regulate gene expression [35]. For example, the MBL protein and circ-MBL are generated from the same gene locus, and circ-MBL can interact with the MBL protein to regulate the balance between MBL protein and circ-MBL [36]. Circ-Foxo3 interacts with both p21 and CDK2 (cycle-dependent kinase 2) to form a circ-Foxo3-p21-CDK2 ternary complex, which inhibits the cell cycle by inhibiting the binding of CDK2 and cyclin E and enhancing the binding of CDK2 and p21 [37].
Regulating splicing and transcription
The majority of ciRNAs and EIciRNAs are retained in the nucleus and can regulate gene expression. The ciRNA ci-ANKRD52 can promote the transcription of its parental gene by affecting Pol II elongation [5]. Circ-EIF3J and circ-PAIP2 are EIciRNAs that facilitate the expression of their parental genes in HeLa and HEK 293 cells by binding to the U1 small nuclear ribonucleic proteins (snRNPs) and interacting with RNA Pol II at the promoter regions of their parental genes [17]. Moreover, the biogenesis of circRNAs may be a way to regulate the expression of parental genes. For most circRNAs produced from exons of protein-coding genes, on the one hand, the more an exon is cyclized, the less it appears in the mRNA [38,39], and on the other hand, there is competition for the usage of 5’ and 3’ splice sites between the biogenesis of circRNAs and linear mRNAs [39].
Acting as protein scaffolds
CircRNAs can modulate protein-protein interactions by acting as dynamic scaffolds [35]. Circ-Foxo3 has both p53 and double mouse minute 2 (MDM2) biding sites, and the p53-MDM2 co-IP was affected by circ-Foxo3 overexpression or knockout, so circ-Foxo3 may serve as a scaffold to mediate the formation of the p53-MDM2 complex [40]. These results suggest that circRNAs with both enzyme and substrate binding sites may serve as scaffolds to close the distance between proteins and facilitate protein reactions.
Translating proteins
Although many circRNAs contain the canonical AUG initiation codon of their host gene, initially they were thought to be unable to be translated into proteins because they lack a 5’ cap, which is the machinery and factors necessary for the formation of translation initiation complexes. However, recently, some circRNAs have been found to be translated, which is driven by N6-methyladenosine (m6A) and ribosome entry site (IRES). The consensus m6A motif near the translation start site can drive the protein translation from circRNAs by recruiting the initiation factor eIF4G2 and the m6A reader YTHDF3 from the cytosol into the nucleus and binding to them [41]. The m6A-driven translation of circRNAs can be inhibited by the m6A demethylase FTO and promoted by the adenosine methyltransferase METTL3/14. Additionally, IRES can recruit and bind ribosomes to initiate translation in a cap-independent manner under stress conditions [42]. CircZNF609 [6] and circ-MBL [7] have been found to contain IRES that can bind to polysomes, and polypeptides translated from these molecules have also been verified.
CircRNA-derived pseudogenes
A classic approach for pseudogene production occurs when an mRNA is reverse transcribed into cDNA and the cDNA is inserted into the genome. Pseudogenes produced in this way maintain the same exon sequence as the parental linear mRNA. However, Dong et al. [43] discovered some circRNA-derived pseudogenes in both mice and human genomes, which had an exon-exon linkage in reverse order of their parental genes and might be generated by the same biogenesis mechanism as mRNA-derived pseudogenes.
CircRNAs in hematopoiesis
Hematopoiesis is a strictly regulated process in which hematopoietic stem cells differentiate into blood cells with specific functions and morphologies, and it involves transcription factors [44], miRNA [45], lncRNA [46], TNF [47] and other chemical factors [48]. CircRNAs have been found to be widely expressed in hematopoietic cells and mature blood cells, with expression that can be altered upon differentiation and can be cell-type specific, rendering such circRNAs as potential hematopoietic regulators. For example, circ-BACH1 (exons 2-4) is preferentially expressed in HSCs (hematopoietic stem cells) and MPPs (multipotent progenitors), circ-FNDC3B in NK cells, circ-MYBL1 and circ-SLFN12L in T cells and NK cells, circ-AKT3 and circ CCDC91 in lymphoid cells, and circ-BACH1 (exons 3-4) in monocytes [24]. Surprisingly, circRNAs are more enriched in enucleated cells, platelets, and red blood cells than in other hematopoietic cells [24,31,49]. Alhasan et al. [49] attributed this phenomenon to the fact that mRNAs are more easily degraded than circRNAs and proposed it as a signature of transcriptome degradation. However, platelets are translationally competent and exhibit active splicing upon platelet activation [50-52], and they share only a part of circRNAs with megakaryocytes [24], which leads us to speculate that circRNAs are involved in biological processes in these cells and are not just byproducts of transcriptome degradation. Preusser et al. confirmed this speculation and revealed that platelets could selectively release circRNAs into microvesicles and exosomes as regulatory signals for certain biological processes such as hemostasis, inflammation, and cancer metastasis [31]. Zhang et al. also found that circRNAs may participate in macrophage differentiation and polarization [53]. Additionally, some circRNAs have been discovered to protect bone marrow stromal cells from total body irradiation [54,55]. Cia-cGAS (circular RNA antagonist for cGAS; cGAS: cyclic GMP-AMP synthase) is richly expressed in long-term HSCs (LT-HSCs) [56], and elevated cia-cGAS can bind to cGAS in the nucleus to inhibit cGAS-mediated production of type I IFNs [57-59], thus protecting LT-HSCs from type I IFN-driven exhaustion [60,61]. Broadly and specifically expressed in hematopoietic cells, circRNAs are not the byproducts of transcriptome degradation but are potential biomarkers for hematopoietic cells and regulators of their differentiation, maturation, and functions. However, the mechanisms of how circRNAs regulate hematopoietic cell differentiation, maturation, and functions have yet to be elucidated. The circRNAs involved in hematopoietic cell differentiation, maturation and functions are shown in Figure 3.
CircRNAs in hematological malignancies
CircRNAs in acute myeloid leukemia
A large number of circRNAs specifically expressed in acute myeloid leukemia have been identified with the development of next-generation sequencing technology. Li et al. discovered that 464 circRNAs were differentially expressed with a 2-fold change cut-off in cytogenetically normal AML (CN-AML) compared with healthy controls. Among them, a dramatically downregulated circRNA, hsa_circ_0004277, can differentiate AML patients from the healthy individuals with an area under the receiver operating characteristic (AUC) curve of 0.957 in a cohort of 115 samples. Downregulation of this circRNA was restored after complete remission and was again downregulated at recurrence [26]. Similarly, Chen et al. performed circRNA microarray analysis on five pairs of newly diagnosed AML and iron deficiency anemia (IDA) cases, finding that 282 circRNAs were upregulated and 416 circRNAs were downregulated in AML. Next, the authors validated these findngs in a cohort of 87 AML and 45 IDA patients by qRT-PCR, confirming that circ-ANAPC7 was dramatically upregulated [27]. Likewise, Lei et al. tested samples from three pairs of AML and IDA patients for RNA-seq, but detailed information on the circRNA expression profiles was not provided. Only the circ_0009910 was suggested to be upregulated in AML in a group of 70 AML patients and 70 IDA controls, as determined by qRT-PCR. In addition, increased expression of circ_0009910 was found to be correlated with high-risk and a shorter overall survival rate [30]. In another study, circRNA microarray and genome-wide microarray analysis were performed simultaneously for the first time in 4 matched AML patients with and without extramedullary infiltration (EMI) as well as healthy volunteers. The results showed that 253 circRNAs and 663 genes were upregulated, and 259 circRNAs and 838 genes were downregulated in EMI AML patients compared with non-EMI AML patients. One of the upregulated circRNAs in AML with EMI, hsa_circRNA_0004520, was validated by performing RT-PCR on four pairs of EMI and non-EMI AML samples [28]. In a study by Li et al., 246 upregulated circRNAs and 262 downregulated circRNAs were identified by RNA-seq to be dynamically expressed during ATRA-induced differentiation. Some dysregulated circRNAs, including circ-HIPK2, were selected for validation by qRT-PCR, and the verification results were consistent with the RNA-seq results, indicating that some circRNAs showed dynamic regulation during ATRA-induced differentiation and may play a role in ATRA-induced differentiation [62]. Similarly, by screening the circRNA expression profiles of three matched THP-1 and doxorubicin-resistant THP-1 (THP-1/ADM) cell lines, Shang et al. revealed a total of 49 circRNAs to be significantly aberrantly expressed in the THP-1/ADM cell lines. Among them, 35 circRNAs were upregulated and 14 circRNAs were downregulated. One of the aberrant circRNAs, circ-PAN3, was found to be markedly upregulated in refractory/recurrent AML by examining its expression levels in 20 refractory/recurrent AML and 22 chemo-sensitive patients [63]. In several other papers, researchers have found circRNAs of interest by searching online databases or the literature. One of the circRNAs, circ-VIM, was discovered to be significantly upregulated in de novo AML and could discriminate AML (AUC = 0.741, 95% CI: 0.657-0.825), non-APL (acute promyelocytic leukemia) AML (AUC = 0.740, 95% CI: 0.652-0.825) and CN-AML (AUC = 0.749, 95% CI: 0.645-0.852) patients from controls in a 155 sample cohort. Moreover, circ-VIM was an independent prognostic factor because patients with elevated circ-VIM had shorter leukemia-free survival and overall survival [64]. CircRNA-DLEU2 was confirmed to be upregulated in a cohort of 20 CN-AML patients and 20 healthy controls [29]. The hsa_circ_100290, extracted from the GSE94591 dataset of the GEO database (http://www.ncbi.nlm.nih.gov/geo/) [65], was confirmed to be markedly upregulated in AML patients compared with the healthy controls in a cohort of 60 AML and 60 healthy samples [66]. CircMYBL2 was also identified from the GSE94591 dataset and was validated to be upregulated in FTL3-ITD (FMS-like tyrosine-3-internal tandem duplication) AML (an AML with poor prognosis) compared with non-FLT3-ITD AML patients by qPCR [67]. The expression levels of some circNPM1 variants (including hsa_circ_0075001) in AML samples are higher than in normal samples, and subtypes M0 and M1 share a higher expression of hsa_circ_0075001 than M2, M4 and M5 [68]. F-circRNAs have also been found in AML with aberrant chromosomal translocation; for example, f-circPR has been found in APL patients and APL cell lines with the PML/RARα and f-circM9 has been found in AML patients who carry the MLL/AF9 [14,69]. These results suggest that circRNAs are abundant and specific in AML, illustrating that circRNAs may serve as promising potential diagnostic biomarkers, prognostic biomarkers or monitoring indicators for AML.
In addition to acting as potential biomarkers, circRNAs can also regulate cell proliferation and apoptosis by sponging miRNAs or can participate in other modes of action involved in initiation, progression and drug resistance in AML. For instance, circRNA-DLEU2 has been demonstrated to promote PRKACB expression by inhibiting the expression of miR-496, thereby promoting the proliferation of AML cells in vitro and the formation of AML tumors in vivo [29]. Circ_0009910 has been observed to improve the growth of AML cells by inhibiting miR-20a-5p in vitro and in vivo [30]. In another study, knockdown of hsa_circ_100290 significantly arrested the cell cycle in G1 phase, promoted apoptosis, and increased the expression levels of apoptosis-related proteins, such as cyclin D1, CDK4, BCl-2, and cleaved caspase-3. Furthermore, bioinformatics and luciferase assays confirmed that miR-203 bound both hsa_circ_100290 and Rab10, and hsa_circ_100290 upregulated Rab10 expression by sponging miR-203, therby promoting the proliferation of AML cells and inhibiting apoptosis [66]. Circ-HIPK2, derived from a gene closely associated with the initiation and progression of AML as a transcriptional coactivators in the nucleus [70,71], has been shown to be needed for ATRA-induced differentiation of APL cells, but its overexpression alone could not induce differentiation of APL cells. MiR-124-3p can be targeted by both circ-HIPK2 and CEBPA and can suppress the expression of CEBPA, a critical hematopoietic transcription factor [72]. Overall, circ-HIPK2 might promote the ATRA-induced differentiation of APL cells by serving as a sponge for miR-124-3p to restore the expression of CEBPA [62]. CircPAN3, whose host gene PAN3 is located in amplified chromosome regions in AML patients with a complex karyotype [73], may employ the circPAN3-miR-153-5p/miR-183-5p-XIAP axis to mediate drug resistance by acting as a miR-153-5p sponge [63]. CirMYBL2 has been found to recruit and bind with the RNA-binding protein PTBP1, facilitating FLT3/FLT3-ITD translation by facilitating the interaction of PTBP1 and FLT3/FLT3-ITD, and ultimately promoting FLT3-ITD AML progression. In contrast, knockdown of circMYBL2 can suppress FLT3-ITD AML progression and impair the drug resistance of FLT3-ITD-positive AML patients [67]. The functions of the above circRNAs have been experimentally verified. The roles of the following circRNAs in AML have only been predicted by bioinformatic analysis or literature review. Li et al. used bioinformatics to predict the target miRNAs of the hsa_circ_0004277 and the target genes of the above miRNAs. Furthermore, GO (http://www.geneontology.org) [74] and KEGG (https://www.kegg.jp/) [75] analyses have shown that the target genes of hsa_circ_0004277 constitute signatures involved in various biological processes and signaling pathways, such as the regulation of cellular processes and cell junctions, as well as the MAPK, PI3K-Akt and HIF-1 signaling pathways [26]. Hsa_circ_0075001 has been observed to be inversely correlated with the expression of some genes that are known targets of miR-181 and are involved in the TLR signaling pathway [68]. Furthermore, miR-181 is commonly downregulated in CN-AML [76], and the TLR signaling pathway has been suggested to regulate the differentiation of leukemic stem cells [77,78], so hsa_circ_0075001 may function via a miR-181 family/TLR signaling pathway axis. Circ-ANAPC was predicted by web-based tools to contain miRNA response elements (MREs) for the hsa-miR-181 family. A KEGG pathway enrichment analysis showed that the hsa-miR-181 family was strictly related to the cancer-related pathway; therefore, circ-ANAPC7 was speculated to be involved in AML tumorigenesis by sponging the miR-181 family [27]. In another study, Lv et al. searched out 17 “kernel” circRNAs that were possibly closely related to EMI and further constructed a ceRNA (competing endogenous RNA) network of circRNA/miRNA/mRNA by bioinformatics analysis to confirm that circRNAs had a bearing on EMI via ceRNA networks [28]. A new class of circRNAs, f-circRNAs have also been shown to promote cellular transformation as pro-oncogenic RNAs. Although they alone were not sufficient to trigger tumorigenesis, they could promote the development of leukemia and confer resistance to therapy when combined with other oncogenic stimuli [14]. The circRNAs validated in hematological malignancies are shown in Table 2.
Table 2.
Disease | CircRNA ID | Gene symbol | Regulation | Biomarker | Role | Potential mechanism | Ref | |
---|---|---|---|---|---|---|---|---|
| ||||||||
Diagnosis | Prognosis | |||||||
AML | circ_0004277 | WDR37 | ↓ | + | - | miR-138-5p/SH3GL2? | [26] | |
circ-ANAPC7 | ANAPC7 | ↑ | + | - | miR-181 family | [27] | ||
circ_0009910 | MFN2 | ↑ | + | + | oncogene | miR-20a-5p | [30] | |
circ_0004520 | VAV2 | ↑ | + | - | miRNA? | [28] | ||
circ_0100181 (circPAN3) | PAN3 | ↑ | - | - | chemoresistance | miR-153-5p,miR-183-5p/XIAP | [63] | |
circ-HIPK2 | ↑ | + | - | facilitate ATRA-inducing differentiation | miR-124a/CEBPA | [62] | ||
circ-VIM | VIM | ↑ | + | + | [64] | |||
circ_0000488 (circRNA-DLEU2) | DLEU2 | ↑ | + | - | oncogene | miR-496/PRKACB | [29] | |
circ_100290 | SLC30A7 | ↑ | + | - | oncogene | miR203/Rab10 | [66] | |
circMYLB2 | MYLB2 | ↑ | + | + | oncogene | PTBP1/FLT3 | [67] | |
circ_0075001 | NPM1 | ↑ | - | - | oncogene? | miR-181/TLR signaling pathway? | [68] | |
circPR | PML-RARα | - | + | - | oncogene, chemoresistance | [14] | ||
circMF9 | MLL/AF9 | - | + | - | oncogene, chemoresistance | [14] | ||
ALL | circ-PVT1 | PVT1 | ↑ | + | - | oncogene | miR-let7/c-MYC, miR-125/Bcl-2? | [86] |
CML | circ_100053 | ↑ | + | + | chemoresistance | [82] | ||
circ_0080145 | TNS3 | ↑ | + | - | oncogene | miR-129b | [83] | |
circBA9.3 | BCR-ABL | ↑ | + | + | oncogene, chemoresistance | upregulate protein c-ABL1 and BCR-ABL1 | [81] | |
CLL | circ_000070 (circ-CBFB1) | CBFB | ↑ | + | + | oncogene | miR-607/FZD/Wntβ signaling pathway | [90] |
circ_0132266 | MTO1 | ↓ | - | - | tumor suppressor | miR-337-3p/PML | [91] | |
lymphoma | circ_101303 (circ_LAMP1) | LAMP1 | ↑ | + | - | oncogene | miR-615-5p/DDR2 | [93] |
MM | circ_000190 | ↓ | + | + | tumor suppressor | miR-767-5p/MAPK | [97] |
“↑” Upregulation; “↓” Downregulation; “+” Yes; “-” No; “blank” Unknown; “?” Not experimentally validated.
In conclusion, circRNAs may be associated with the initiation, progression, and drug resistance of AML through multiple action modes, indicating that they are the potential therapeutic targets.
CircRNAs in chronic myeloid leukemia
Chronic myeloid leukemia (CML) is a clonal disease of hematopoietic stem cells, characterized by the aberrant chromosome translocation of t (9;22), resulting in fusion of the BCR-ABL1 gene and leading to sustained tyrosine kinase activity [79]. In the management of CML, the detection of BCR-ABL mRNA by qRT-PCR is the “gold standard” for the evaluation of therapeutic effect and prediction of recurrence [80].
Recently, Pan et al. discovered a fusion circRNA, circBA9.3, that can promote cell proliferation and endow leukemic cells with resistance to TKIs (trosine kinase inhibitors, including imatinib, nilotinib, and dasatinib) by upregulating c-ABL1 and BCR-ABL1 protein expression levels instead of the BCR-ABL1 oncogene [81]. CircBA9.3 is generated from the fusion gene BCR-ABL1 of CML patients, which is markedly overexpressed in TKI-resistant CML patients compared with TKI-susceptible CML patients. Another circRNA, hsa_circ_100053, was shown to be remarkably increased in CML, which was verified by qRT-PCR of 151 CML and 100 healthy samples [82]. The expression level of hsa_circ_100053 has been related to the relative risk of Sokal (P = 0.037) and the mutation status of BCR/ABL (P = 0.008). Moreover, the high expression of hsa_circ_100053 was not only an independent prognostic factor for CML progression (P = 0.037), indicating a shorter overall survival, but also conferred drug resistance to CML patients and cell lines. In another study, Liu et al. identified 361 differentially expressed circRNAs between CML and control samples with a fold change threshold of >2, including 201 upregulated and 160 downregulated circRNAs [83]. Among them, the upregulation of hsa_circ_0080145 in CML patients and cell lines was validated by qRT-PCR. Subsequently, the finding that hsa_circ_0080145 could promote CML proliferation by serving as a sponge of miR-29b and could play an oncogenic role in the progression of CML was confirmed through dual luciferase assay and a series of loss-of-function and rescue experiments.
These studies have demonstrated that circRNAs are differentially expressed in CML, are potential diagnostic or prognostic biomarkers and even participate in the initiation, progression and drug resistance of CML. Most importantly, the result that circBA9.3 promotes cell proliferation and endows CML with TKI resistance by regulating c-ABL1 and BCR-ABL1 protein expression rather than BCR-ABL1 oncogene expression challenges the use of BCR-ABL1 mRNA detection in traditional CML patient management.
CircRNAs in lymphoid malignancies
Lymphoid malignancies are a group of clonal tumors that arise at different stages of B/T-cell development. Here we mainly focused on acute lymphoblastic leukemia (ALL), chronic lymphoblastic leukemia (CLL), lymphoma, and multiple myeloma (MM).
Acute lymphoblastic leukemia (ALL) is a heterogeneous group of lymphoid malignancies caused by the monoclonal proliferation and expansion of B/T precursor lymphoid cells in the bone marrow, blood and other organs [84]. With treatment, pediatric ALL has a cure rate of approximately 90% [85]. However, treatment of adults is unsatisfactory and full of challenges. Elucidating the pathogenesis of ALL is the primary task of research on ALL. Hu et al. showed that circ-PVT1 is produced from the long noncoding RNA PVT1 locus on chromosome 8q23, is mainly located in the cytosol and may play an oncogenic role in ALL [86]. Circ-PVT1 was strikingly elevated in ALL cell lines and ALL patient samples. In-depth functional experiments have demonstrated that circ-PVT1 knockdown can inhibit cell proliferation and induce apoptosis, accompanied by decreased levels of the c-MYC and Bcl-2 proteins. Circ-PVT1 can act as a sponge for miRNA let-7 [87] and miRNA-125 [88], and in turn, these microRNAs target c-MYC and Bcl-2, respectively. Therefore, knockdown of circPVT1 can arrest cell proliferation and promote apoptosis possibly by elevating miRNA let-7 and miRNA-125 levels to suppress the expression of c-MYC and Bcl-2 in ALL cell lines. However, this hypothesis has not been confirmed by further experiments.
Chronic lymphoblastic leukemia (CLL), the type of leukemia with the highest incidence in western countries, is a hematopoietic malignancy with clonal proliferation and accumulation of mature CD5-positive B-cells [89]. Although patient survival has been improved by introducing fludarabine, an anti-20 monoclonal antibody, and Bruton’s tyrosine kinase, the pathogenesis of CLL still needs further study because of its recurrence. Recently, a study provided new insight into the pathogenesis of CLL from the perspective of ncRNA, indicating that circ-CBFB generated from chromosome 1 was dramatically upregulated in blood samples from CLL patients [90]. Circ-CBFB was suggested to distinguish CLL patients from healthy controls with an AUC of 0.80 (95% CI: 0.69-0.90). High circ-CBFB expression was an independent predictor of prognosis and was associated with shorter survival time. Furthermore, circ-CBFB was shown to be an oncogenic stimulus for CLL through the circ-CBFB/miR-607/FZD/Wnt/β-catenin signaling pathway axis by serving as a miRNA sponge. In another study, Wu et al. revealed that circ_0132266 was dramatically downregulated in CLL patients compared with healthy controls and exerted a tumor-suppressive role in CLL by the miR-337-3p/PML axis, playing a role as a miRNA sponge [91].
Lymphoma is a heterogeneous lymphoid malignancy. Its clinical manifestations, therapeutic responses and prognosis vary with the histological type, clinical factors and molecular characteristics [92]. Deng et al. discovered that circ-LAMP1 was dramatically overexpressed in T-cell lymphoblastic lymphoma tissues compared with normal infantile thymus and may play an oncogenic role in T-cell lymphoblastic lymphoma because knockdown of circ-LAMP1 inhibited cell viability and dramatically increased the rate of apoptosis, and vice versa [93]. In-depth mechanistic studies have revealed that circ-LAMP1 promotes DDR2 expression and regulates cell proliferation and apoptosis in T-cell lymphoblastic lymphoma by sponging miR-615-5p. Deng et al. also stated that it was challenging to detect RNA in frozen samples. Dahl et al. encountered the same problem, and they presented a more accurate and sensitive circRNA quantification method, which they named NanoString assay [94]. The authors employed RNA-seq to screen the circRNA expression profiles of four different mantle cell lymphoma (MCL) cell lines and one multiple myeloma (MM) cell line and they discovered 813, 816, 741, 279 and 619 unique circRNAs, respectively. Then, they verified the structures and abnormal expression of some of the circRNAs by Sanger sequencing analysis, qRT-PCR and northern blotting analysis and found that the circRNA expression profiles could distinguish different B-cell malignancies by Hierarchical Cluster Analysis [95], which indicated that circRNAs might act as potential diagnostic and differential diagnostic biomarkers. Importantly, this study showed that the NanoString technology is superior to RNA-seq and qRT-PCR in sensitivity, specificity, and quantitative accuracy and is a better method for detecting circRNAs because it dose not require RT (reverse transcription) and PCR amplification, which is a major source of assay artifacts. NanoString technology is also reliable, even in low-quality RNA samples such as formalin-fixed and paraffin-embedded tissues.
Multiple myeloma (MM) is a highly heterogeneous disease characterized by the accumulation of clonal plasma in the bone marrow [96]. In the latest study, circ_0000190 was shown to be downregulated not only in MM cell lines but also in the bone marrow and plasm of MM patients compared with healthy controls [97]. International staging system stage I and Durie-Salmon stage I patients had higher circ_0000190 levels than stage II and III patients, and higher expression of circ_0000190 was positively correlated with a longer progression-free period and overall survival time, indicating that higher expression levels of circ_0000190 predicted excellent clinical outcome. In-depth research demonstrated that circ_0000190 inhibited miR-767-5p and then upregulated MAPK levels, ultimately arresting the cell cycle and inhibiting MM progression. Additionally, circ_0000190 was shown to inhibit tumor progression in an MM mouse model.
These studies revealed that circRNAs are broadly and specifically expressed in various subtypes of lymphoid malignancies as they are in other diseases, indicating that circRNAs can also be diagnostic biomarkers for lymphoid malignancies. Additionally, some of circRNAs have been associated with prognosis, some with disease occurrence and progression, and some may have antitumor effects.
Conclusions
In this review, the biogenesis, categories, functions, and characteristics of circRNAs were summarized, with emphasis on the roles of circRNAs in the hematological system. CircRNAs are widely and specifically expressed in hematopoietic stem cells, mature blood cells and various subtypes of hematological malignancies and can be detected in blood and bone marrow, making them promising potential diagnostic biomarkers. The expression of some circRNAs is associated with the current cancer risk stratification system, progression-free survival time, overall survival time, extramedullary infiltration and drug resistance, suggesting that they may be potential prognostic biomarkers and monitoring indicators. Some circRNAs are dynamically expressed during hematopoietic differentiation, some can protect mesenchymal stem cells from total body irradiation and some can maintain the self-renewal and differentiation ability of stem cells by sponging miRNAs or acting with other molecules, which suggests that circRNAs may be potential hematopoietic cell biomarkers and hematopoietic regulators. Additionally, some circRNAs have been confirmed to act with miRNAs or RBPs to regulate the expression of downstream genes and eventually participate in the occurrence and development of hematological malignancies as well as in drug resistance by influencing the cell cycle, signaling pathways and posttranscriptional regulation, which indicates that circRNAs can be potential therapeutic targets.
Although we have gradually unveiled the mystery of circRNAs, our understanding of circRNAs may be just the tip of the iceberg and still far from circRNA clinical application. Here, we also include some suggestions for future research on circRNAs in hematological malignancies. (1) It is advisable to provide the names of circRNAs cataloged in the circBase database (http://www.circbase.org/) [98], which is the most complete database for circRNAs annotation at present. The naming of circRNAs is currently very confusing, which makes it difficult for researchers to communicate with each other. (2) Although many unique circRNAs in hematologic malignancies have been identified by RNA-seq or circRNA microarrays, only a small portion of these circRNAs have been verified in a large number of samples, and multiple circRNAs remain to be investigated. (3) Many circRNAs have been identified to be differentially expressed in different stages of hematopoietic cells, but how they regulate differentiation, maturation and functions are still not clear, so the mechanism of circRNAs as hematopoietic regulators needs to be addressed in future studies. (4) Increasing circRNAs have been confirmed as diagnostic markers, but their sensitivity and specificity have yet to be verified. Combining several circRNAs or combining circRNAs with traditional diagnostic biomarkers may improve the diagnostic value. (5) Most studies on circRNAs in hematological malignancies have focused on the role of circRNAs as miRNA sponges. On the one hand, the exact mechanisms remain to be further studied, and on the other hand, other action modes of circRNAs are also worth exploring. (6) Some circRNAs have been confirmed to be sensitive to RNase, and it can be challenging to extract circRNAs from some low-quality specimens. More sensitive and accurate circRNA detection methods need to be developed. In general, research on circRNAs is still in its infancy, but with the development of the next-generation high-throughput sequencing technology, bioinformatic analysis and the use of online databases, we can expect more circRNAs and their action modes to be elucidated and applied to prevent and treat hematological malignancies.
Acknowledgements
This work was supported by grants from the Medical and Health Science and Technology Projects of Zhejiang Province (2019KY171), the National Science Foundation of Ningbo (2018A610390), the National Science Foundation of Zhejiang Province (LY17H160005), and the Chinese Medicine Science and Technology Plan Project of Zhejiang Province (2015ZZ018).
Disclosure of conflict of interest
None.
References
- 1.Sanger HL, Klotz G, Riesner D, Gross HJ, Kleinschmidt AK. Viroids are single-stranded covalently closed circular RNA molecules existing as highly base-paired rod-like structures. Proc Natl Acad Sci U S A. 1976;73:3852–3856. doi: 10.1073/pnas.73.11.3852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Hsu MT, Coca-Prados M. Electron microscopic evidence for the circular form of RNA in the cytoplasm of eukaryotic cells. Nature. 1979;280:339–340. doi: 10.1038/280339a0. [DOI] [PubMed] [Google Scholar]
- 3.Zhang J, Liu H, Hou L, Wang G, Zhang R, Huang Y, Chen X, Zhu J. Circular RNA_LARP4 inhibits cell proliferation and invasion of gastric cancer by sponging miR-424-5p and regulating LATS1 expression. Mol Cancer. 2017;16:151. doi: 10.1186/s12943-017-0719-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Chen S, Huang V, Xu X, Livingstone J, Soares F, Jeon J, Zeng Y, Hua JT, Petricca J, Guo H, Wang M, Yousif F, Zhang Y, Donmez N, Ahmed M, Volik S, Lapuk A, Chua MLK, Heisler LE, Foucal A, Fox NS, Fraser M, Bhandari V, Shiah YJ, Guan J, Li J, Orain M, Picard V, Hovington H, Bergeron A, Lacombe L, Fradet Y, Tetu B, Liu S, Feng F, Wu X, Shao YW, Komor MA, Sahinalp C, Collins C, Hoogstrate Y, de Jong M, Fijneman RJA, Fei T, Jenster G, van der Kwast T, Bristow RG, Boutros PC, He HH. Widespread and functional RNA circularization in localized prostate cancer. Cell. 2019;176:831–843. e822. doi: 10.1016/j.cell.2019.01.025. [DOI] [PubMed] [Google Scholar]
- 5.Zhang Y, Zhang XO, Chen T, Xiang JF, Yin QF, Xing YH, Zhu S, Yang L, Chen LL. Circular intronic long noncoding RNAs. Mol Cell. 2013;51:792–806. doi: 10.1016/j.molcel.2013.08.017. [DOI] [PubMed] [Google Scholar]
- 6.Legnini I, Di Timoteo G, Rossi F, Morlando M, Briganti F, Sthandier O, Fatica A, Santini T, Andronache A, Wade M, Laneve P, Rajewsky N, Bozzoni I. Circ-ZNF609 is a circular RNA that can be translated and functions in myogenesis. Mol Cell. 2017;66:22–37. e29. doi: 10.1016/j.molcel.2017.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Pamudurti NR, Bartok O, Jens M, Ashwal-Fluss R, Stottmeister C, Ruhe L, Hanan M, Wyler E, Perez-Hernandez D, Ramberger E, Shenzis S, Samson M, Dittmar G, Landthaler M, Chekulaeva M, Rajewsky N, Kadener S. Translation of CircRNAs. Mol Cell. 2017;66:9–21. e27. doi: 10.1016/j.molcel.2017.02.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bonizzato A, Gaffo E, Te Kronnie G, Bortoluzzi S. CircRNAs in hematopoiesis and hematological malignancies. Blood Cancer J. 2016;6:e483. doi: 10.1038/bcj.2016.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Liu Y, Cheng Z, Pang Y, Cui L, Qian T, Quan L, Zhao H, Shi J, Ke X, Fu L. Role of microRNAs, circRNAs and long noncoding RNAs in acute myeloid leukemia. J Hematol Oncol. 2019;12:51. doi: 10.1186/s13045-019-0734-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Li X, Yang L, Chen LL. The biogenesis, functions, and challenges of circular RNAs. Mol Cell. 2018;71:428–442. doi: 10.1016/j.molcel.2018.06.034. [DOI] [PubMed] [Google Scholar]
- 11.Noto JJ, Schmidt CA, Matera AG. Engineering and expressing circular RNAs via tRNA splicing. RNA Biol. 2017;14:978–984. doi: 10.1080/15476286.2017.1317911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Schmidt CA, Noto JJ, Filonov GS, Matera AG. A method for expressing and imaging abundant, stable, circular RNAs in vivo using tRNA splicing. Methods Enzymol. 2016;572:215–236. doi: 10.1016/bs.mie.2016.02.018. [DOI] [PubMed] [Google Scholar]
- 13.Lu Z, Filonov GS, Noto JJ, Schmidt CA, Hatkevich TL, Wen Y, Jaffrey SR, Matera AG. Metazoan tRNA introns generate stable circular RNAs in vivo. RNA. 2015;21:1554–1565. doi: 10.1261/rna.052944.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Guarnerio J, Bezzi M, Jeong JC, Paffenholz SV, Berry K, Naldini MM, Lo-Coco F, Tay Y, Beck AH, Pandolfi PP. Oncogenic role of fusion-circRNAs derived from cancer-associated chromosomal translocations. Cell. 2016;165:289–302. doi: 10.1016/j.cell.2016.03.020. [DOI] [PubMed] [Google Scholar]
- 15.Liang D, Tatomer DC, Luo Z, Wu H, Yang L, Chen LL, Cherry S, Wilusz JE. The output of protein-coding genes shifts to circular RNAs when the Pre-mRNA processing machinery is limiting. Mol Cell. 2017;68:940–954. e943. doi: 10.1016/j.molcel.2017.10.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Vo JN, Cieslik M, Zhang Y, Shukla S, Xiao L, Zhang Y, Wu YM, Dhanasekaran SM, Engelke CG, Cao X, Robinson DR, Nesvizhskii AI, Chinnaiyan AM. The landscape of circular RNA in cancer. Cell. 2019;176:869–881. e813. doi: 10.1016/j.cell.2018.12.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, Zhong G, Yu B, Hu W, Dai L, Zhu P, Chang Z, Wu Q, Zhao Y, Jia Y, Xu P, Liu H, Shan G. Exon-intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol. 2015;22:256–264. doi: 10.1038/nsmb.2959. [DOI] [PubMed] [Google Scholar]
- 18.Jeck WR, Sorrentino JA, Wang K, Slevin MK, Burd CE, Liu J, Marzluff WF, Sharpless NE. Circular RNAs are abundant, conserved, and associated with ALU repeats. RNA. 2013;19:141–157. doi: 10.1261/rna.035667.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Conn SJ, Pillman KA, Toubia J, Conn VM, Salmanidis M, Phillips CA, Roslan S, Schreiber AW, Gregory PA, Goodall GJ. The RNA binding protein quaking regulates formation of circRNAs. Cell. 2015;160:1125–1134. doi: 10.1016/j.cell.2015.02.014. [DOI] [PubMed] [Google Scholar]
- 20.Gao Y, Wang J, Zhao F. CIRI: an efficient and unbiased algorithm for de novo circular RNA identification. Genome Biol. 2015;16:4. doi: 10.1186/s13059-014-0571-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Suzuki H, Tsukahara T. A view of pre-mRNA splicing from RNase R resistant RNAs. Int J Mol Sci. 2014;15:9331–9342. doi: 10.3390/ijms15069331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Li P, Chen S, Chen H, Mo X, Li T, Shao Y, Xiao B, Guo J. Using circular RNA as a novel type of biomarker in the screening of gastric cancer. Clin Chim Acta. 2015;444:132–136. doi: 10.1016/j.cca.2015.02.018. [DOI] [PubMed] [Google Scholar]
- 23.Barrett SP, Salzman J. Circular RNAs: analysis, expression and potential functions. Development. 2016;143:1838–1847. doi: 10.1242/dev.128074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Nicolet BP, Engels S, Aglialoro F, van den Akker E, von Lindern M, Wolkers MC. Circular RNA expression in human hematopoietic cells is widespread and cell-type specific. Nucleic Acids Res. 2018;46:8168–8180. doi: 10.1093/nar/gky721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.AbouHaidar MG, Venkataraman S, Golshani A, Liu B, Ahmad T. Novel coding, translation, and gene expression of a replicating covalently closed circular RNA of 220 nt. Proc Natl Acad Sci U S A. 2014;111:14542–14547. doi: 10.1073/pnas.1402814111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Li W, Zhong C, Jiao J, Li P, Cui B, Ji C, Ma D. Characterization of hsa_circ_0004277 as a new biomarker for acute myeloid leukemia via circular RNA profile and bioinformatics analysis. Int J Mol Sci. 2017;18 doi: 10.3390/ijms18030597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Chen H, Liu T, Liu J, Feng Y, Wang B, Wang J, Bai J, Zhao W, Shen Y, Wang X, Yang J, Ji Y, He A, Yang Y. Circ-ANAPC7 is upregulated in acute myeloid leukemia and appears to target the MiR-181 family. Cell Physiol Biochem. 2018;47:1998–2007. doi: 10.1159/000491468. [DOI] [PubMed] [Google Scholar]
- 28.Lv C, Sun L, Guo Z, Li H, Kong D, Xu B, Lin L, Liu T, Guo D, Zhou J, Li Y. Circular RNA regulatory network reveals cell-cell crosstalk in acute myeloid leukemia extramedullary infiltration. J Transl Med. 2018;16:361. doi: 10.1186/s12967-018-1726-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Wu DM, Wen X, Han XR, Wang S, Wang YJ, Shen M, Fan SH, Zhang ZF, Shan Q, Li MQ, Hu B, Chen GQ, Lu J, Zheng YL. Role of circular RNA DLEU2 in human acute myeloid leukemia. Mol Cell Biol. 2018;38 doi: 10.1128/MCB.00259-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ping L, Jian-Jun C, Chu-Shu L, Guang-Hua L, Ming Z. Silencing of circ_0009910 inhibits acute myeloid leukemia cell growth through increasing miR-20a-5p. Blood Cells Mol Dis. 2019;75:41–47. doi: 10.1016/j.bcmd.2018.12.006. [DOI] [PubMed] [Google Scholar]
- 31.Preusser C, Hung LH, Schneider T, Schreiner S, Hardt M, Moebus A, Santoso S, Bindereif A. Selective release of circRNAs in platelet-derived extracellular vesicles. J Extracell Vesicles. 2018;7:1424473. doi: 10.1080/20013078.2018.1424473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK, Kjems J. Natural RNA circles function as efficient microRNA sponges. Nature. 2013;495:384–388. doi: 10.1038/nature11993. [DOI] [PubMed] [Google Scholar]
- 33.Hansen TB, Wiklund ED, Bramsen JB, Villadsen SB, Statham AL, Clark SJ, Kjems J. miRNA-dependent gene silencing involving Ago2-mediated cleavage of a circular antisense RNA. EMBO J. 2011;30:4414–22. doi: 10.1038/emboj.2011.359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Piwecka M, Glazar P, Hernandez-Miranda LR, Memczak S, Wolf SA, Rybak-Wolf A, Filipchyk A, Klironomos F, Cerda Jara CA, Fenske P, Trimbuch T, Zywitza V, Plass M, Schreyer L, Ayoub S, Kocks C, Kuhn R, Rosenmund C, Birchmeier C, Rajewsky N. Loss of a mammalian circular RNA locus causes miRNA deregulation and affects brain function. Science. 2017;357 doi: 10.1126/science.aam8526. [DOI] [PubMed] [Google Scholar]
- 35.Du WW, Zhang C, Yang W, Yong T, Awan FM, Yang BB. Identifying and Characterizing circRNA-Protein Interaction. Theranostics. 2017;7:4183–4191. doi: 10.7150/thno.21299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ashwal-Fluss R, Meyer M, Pamudurti NR, Ivanov A, Bartok O, Hanan M, Evantal N, Memczak S, Rajewsky N, Kadener S. circRNA biogenesis competes with pre-mRNA splicing. Mol Cell. 2014;56:55–66. doi: 10.1016/j.molcel.2014.08.019. [DOI] [PubMed] [Google Scholar]
- 37.Du WW, Yang W, Liu E, Yang Z, Dhaliwal P, Yang BB. Foxo3 circular RNA retards cell cycle progression via forming ternary complexes with p21 and CDK2. Nucleic Acids Res. 2016;44:2846–2858. doi: 10.1093/nar/gkw027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zhang XO, Wang HB, Zhang Y, Lu X, Chen LL, Yang L. Complementary sequence-mediated exon circularization. Cell. 2014;159:134–147. doi: 10.1016/j.cell.2014.09.001. [DOI] [PubMed] [Google Scholar]
- 39.Kelly S, Greenman C, Cook PR, Papantonis A. Exon skipping is correlated with exon circularization. J Mol Biol. 2015;427:2414–2417. doi: 10.1016/j.jmb.2015.02.018. [DOI] [PubMed] [Google Scholar]
- 40.Du WW, Fang L, Yang W, Wu N, Awan FM, Yang Z, Yang BB. Induction of tumor apoptosis through a circular RNA enhancing Foxo3 activity. Cell Death Differ. 2017;24:357–370. doi: 10.1038/cdd.2016.133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Yang Y, Fan X, Mao M, Song X, Wu P, Zhang Y, Jin Y, Yang Y, Chen LL, Wang Y, Wong CC, Xiao X, Wang Z. Extensive translation of circular RNAs driven by N(6)-methyladenosine. Cell Res. 2017;27:626–641. doi: 10.1038/cr.2017.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Jackson RJ, Hellen CU, Pestova TV. The mechanism of eukaryotic translation initiation and principles of its regulation. Nat Rev Mol Cell Biol. 2010;11:113–127. doi: 10.1038/nrm2838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Dong R, Zhang XO, Zhang Y, Ma XK, Chen LL, Yang L. CircRNA-derived pseudogenes. Cell Res. 2016;26:747–750. doi: 10.1038/cr.2016.42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Goode DK, Obier N, Vijayabaskar MS, Lie-A-Ling M, Lilly AJ, Hannah R, Lichtinger M, Batta K, Florkowska M, Patel R, Challinor M, Wallace K, Gilmour J, Assi SA, Cauchy P, Hoogenkamp M, Westhead DR, Lacaud G, Kouskoff V, Gottgens B, Bonifer C. Dynamic gene regulatory networks drive hematopoietic specification and differentiation. Dev Cell. 2016;36:572–587. doi: 10.1016/j.devcel.2016.01.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Bissels U, Bosio A, Wagner W. MicroRNAs are shaping the hematopoietic landscape. Haematologica. 2012;97:160–167. doi: 10.3324/haematol.2011.051730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Luo M, Jeong M, Sun D, Park HJ, Rodriguez BA, Xia Z, Yang L, Zhang X, Sheng K, Darlington GJ, Li W, Goodell MA. Long non-coding RNAs control hematopoietic stem cell function. Cell Stem Cell. 2015;16:426–438. doi: 10.1016/j.stem.2015.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Pronk CJ, Veiby OP, Bryder D, Jacobsen SE. Tumor necrosis factor restricts hematopoietic stem cell activity in mice: involvement of two distinct receptors. J Exp Med. 2011;208:1563–1570. doi: 10.1084/jem.20110752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Bruns I, Lucas D, Pinho S, Ahmed J, Lambert MP, Kunisaki Y, Scheiermann C, Schiff L, Poncz M, Bergman A, Frenette PS. Megakaryocytes regulate hematopoietic stem cell quiescence through CXCL4 secretion. Nat Med. 2014;20:1315–1320. doi: 10.1038/nm.3707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Alhasan AA, Izuogu OG, Al-Balool HH, Steyn JS, Evans A, Colzani M, Ghevaert C, Mountford JC, Marenah L, Elliott DJ, Santibanez-Koref M, Jackson MS. Circular RNA enrichment in platelets is a signature of transcriptome degradation. Blood. 2016;127:e1–e11. doi: 10.1182/blood-2015-06-649434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Santoso S, Kalb R, Kiefel V, Mueller-Eckhardt C. The presence of messenger RNA for HLA class I in human platelets and its capability for protein biosynthesis. Br J Haematol. 1993;84:451–456. doi: 10.1111/j.1365-2141.1993.tb03100.x. [DOI] [PubMed] [Google Scholar]
- 51.Lindemann S, Tolley ND, Eyre JR, Kraiss LW, Mahoney TM, Weyrich AS. Integrins regulate the intracellular distribution of eukaryotic initiation factor 4E in platelets. A checkpoint for translational control. J Biol Chem. 2001;276:33947–33951. doi: 10.1074/jbc.M104281200. [DOI] [PubMed] [Google Scholar]
- 52.Denis MM, Tolley ND, Bunting M, Schwertz H, Jiang H, Lindemann S, Yost CC, Rubner FJ, Albertine KH, Swoboda KJ, Fratto CM, Tolley E, Kraiss LW, McIntyre TM, Zimmerman GA, Weyrich AS. Escaping the nuclear confines: signal-dependent pre-mRNA splicing in anucleate platelets. Cell. 2005;122:379–391. doi: 10.1016/j.cell.2005.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Zhang Y, Zhang Y, Li X, Zhang M, Lv K. Microarray analysis of circular RNA expression patterns in polarized macrophages. Int J Mol Med. 2017;39:373–379. doi: 10.3892/ijmm.2017.2852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wang Y, Zhang J, Li J, Gui R, Nie X, Huang R. CircRNA_014511 affects the radiosensitivity of bone marrow mesenchymal stem cells by binding to miR-29b-2-5p. Bosn J Basic Med Sci. 2019;19:155–163. doi: 10.17305/bjbms.2019.3935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zhang J, Jiang J, Huang R, Wang Y, Nie X, Gui R. Circular RNA expression profiles are significantly altered in mice bone marrow stromal cells after total body irradiation. Leuk Res. 2018;70:67–73. doi: 10.1016/j.leukres.2018.05.010. [DOI] [PubMed] [Google Scholar]
- 56.Xia P, Wang S, Ye B, Du Y, Li C, Xiong Z, Qu Y, Fan Z. A circular RNA protects dormant hematopoietic stem cells from DNA sensor cGAS-mediated exhaustion. Immunity. 2018;48:688–701. e687. doi: 10.1016/j.immuni.2018.03.016. [DOI] [PubMed] [Google Scholar]
- 57.Chen Q, Sun L, Chen ZJ. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat Immunol. 2016;17:1142–1149. doi: 10.1038/ni.3558. [DOI] [PubMed] [Google Scholar]
- 58.Hornung V, Hartmann R, Ablasser A, Hopfner KP. OAS proteins and cGAS: unifying concepts in sensing and responding to cytosolic nucleic acids. Nat Rev Immunol. 2014;14:521–528. doi: 10.1038/nri3719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Xia P, Wang S, Gao P, Gao G, Fan Z. DNA sensor cGAS-mediated immune recognition. Protein Cell. 2016;7:777–791. doi: 10.1007/s13238-016-0320-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Essers MA, Offner S, Blanco-Bose WE, Waibler Z, Kalinke U, Duchosal MA, Trumpp A. IFNalpha activates dormant haematopoietic stem cells in vivo. Nature. 2009;458:904–908. doi: 10.1038/nature07815. [DOI] [PubMed] [Google Scholar]
- 61.Sato T, Onai N, Yoshihara H, Arai F, Suda T, Ohteki T. Interferon regulatory factor-2 protects quiescent hematopoietic stem cells from type I interferon-dependent exhaustion. Nat Med. 2009;15:696–700. doi: 10.1038/nm.1973. [DOI] [PubMed] [Google Scholar]
- 62.Li S, Ma Y, Tan Y, Ma X, Zhao M, Chen B, Zhang R, Chen Z, Wang K. Profiling and functional analysis of circular RNAs in acute promyelocytic leukemia and their dynamic regulation during all-trans retinoic acid treatment. Cell Death Dis. 2018;9:651. doi: 10.1038/s41419-018-0699-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Shang J, Chen WM, Wang ZH, Wei TN, Chen ZZ, Wu WB. CircPAN3 mediates drug resistance in acute myeloid leukemia through the miR-153-5p/miR-183-5p-XIAP axis. Exp Hematol. 2019;70:42–54. e3. doi: 10.1016/j.exphem.2018.10.011. [DOI] [PubMed] [Google Scholar]
- 64.Yi YY, Yi J, Zhu X, Zhang J, Zhou J, Tang X, Lin J, Wang P, Deng ZQ. Circular RNA of vimentin expression as a valuable predictor for acute myeloid leukemia development and prognosis. J Cell Physiol. 2019;234:3711–3719. doi: 10.1002/jcp.27145. [DOI] [PubMed] [Google Scholar]
- 65.Edgar R, Domrachev M, Lash AE. Gene expression omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30:207–210. doi: 10.1093/nar/30.1.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Fan H, Li Y, Liu C, Liu Y, Bai J, Li W. Circular RNA-100290 promotes cell proliferation and inhibits apoptosis in acute myeloid leukemia cells via sponging miR-203. Biochem Biophys Res Commun. 2018;507:178–184. doi: 10.1016/j.bbrc.2018.11.002. [DOI] [PubMed] [Google Scholar]
- 67.Sun YM, Wang WT, Zeng ZC, Chen TQ, Han C, Pan Q, Huang W, Fang K, Sun LY, Zhou YF, Luo XQ, Luo C, Du X, Chen YQ. CircMYBL2, a circRNA from MYBL2, regulates FLT3 translation by recruiting PTBP1 to promote FLT3-ITD AML progression. Blood. 2019 doi: 10.1182/blood.2019000802. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Hirsch S, Blatte TJ, Grasedieck S, Cocciardi S, Rouhi A, Jongen-Lavrencic M, Paschka P, Kronke J, Gaidzik VI, Dohner H, Schlenk RF, Kuchenbauer F, Dohner K, Dolnik A, Bullinger L. Circular RNAs of the nucleophosmin (NPM1) gene in acute myeloid leukemia. Haematologica. 2017;102:2039–2047. doi: 10.3324/haematol.2017.172866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Dal Molin A, Bresolin S, Gaffo E, Tretti C, Boldrin E, Meyer LH, Guglielmelli P, Vannucchi AM, Te Kronnie G, Bortoluzzi S. CircRNAs are here to stay: a perspective on the MLL recombinome. Front Genet. 2019;10:88. doi: 10.3389/fgene.2019.00088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Li XL, Arai Y, Harada H, Shima Y, Yoshida H, Rokudai S, Aikawa Y, Kimura A, Kitabayashi I. Mutations of the HIPK2 gene in acute myeloid leukemia and myelodysplastic syndrome impair AML1- and p53-mediated transcription. Oncogene. 2007;26:7231–7239. doi: 10.1038/sj.onc.1210523. [DOI] [PubMed] [Google Scholar]
- 71.Shima Y, Honma Y, Kitabayashi I. PML-RARalpha and its phosphorylation regulate pml oligomerization and HIPK2 stability. Cancer Res. 2013;73:4278–4288. doi: 10.1158/0008-5472.CAN-12-3814. [DOI] [PubMed] [Google Scholar]
- 72.Hackanson B, Bennett KL, Brena RM, Jiang J, Claus R, Chen SS, Blagitko-Dorfs N, Maharry K, Whitman SP, Schmittgen TD, Lubbert M, Marcucci G, Bloomfield CD, Plass C. Epigenetic modification of CCAAT/enhancer binding protein alpha expression in acute myeloid leukemia. Cancer Res. 2008;68:3142–3151. doi: 10.1158/0008-5472.CAN-08-0483. [DOI] [PubMed] [Google Scholar]
- 73.Mrozek K. Cytogenetic, molecular genetic, and clinical characteristics of acute myeloid leukemia with a complex karyotype. Semin Oncol. 2008;35:365–377. doi: 10.1053/j.seminoncol.2008.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, Harris MA, Hill DP, Issel-Tarver L, Kasarskis A, Lewis S, Matese JC, Richardson JE, Ringwald M, Rubin GM, Sherlock G. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 2000;25:25–29. doi: 10.1038/75556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kanehisa M. A database for post-genome analysis. Trends Genet. 1997;13:375–376. doi: 10.1016/s0168-9525(97)01223-7. [DOI] [PubMed] [Google Scholar]
- 76.Marcucci G, Radmacher MD, Maharry K, Mrozek K, Ruppert AS, Paschka P, Vukosavljevic T, Whitman SP, Baldus CD, Langer C, Liu CG, Carroll AJ, Powell BL, Garzon R, Croce CM, Kolitz JE, Caligiuri MA, Larson RA, Bloomfield CD. MicroRNA expression in cytogenetically normal acute myeloid leukemia. N Engl J Med. 2008;358:1919–1928. doi: 10.1056/NEJMoa074256. [DOI] [PubMed] [Google Scholar]
- 77.Nagai Y, Garrett KP, Ohta S, Bahrun U, Kouro T, Akira S, Takatsu K, Kincade PW. Toll-like receptors on hematopoietic progenitor cells stimulate innate immune system replenishment. Immunity. 2006;24:801–812. doi: 10.1016/j.immuni.2006.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Okamoto M, Hirai H, Taniguchi K, Shimura K, Inaba T, Shimazaki C, Taniwaki M, Imanishi J. Toll-like receptors (TLRs) are expressed by myeloid leukaemia cell lines, but fail to trigger differentiation in response to the respective TLR ligands. Br J Haematol. 2009;147:585–587. doi: 10.1111/j.1365-2141.2009.07858.x. [DOI] [PubMed] [Google Scholar]
- 79.Apperley JF. Chronic myeloid leukaemia. Lancet. 2015;385:1447–1459. doi: 10.1016/S0140-6736(13)62120-0. [DOI] [PubMed] [Google Scholar]
- 80.Baccarani M, Deininger MW, Rosti G, Hochhaus A, Soverini S, Apperley JF, Cervantes F, Clark RE, Cortes JE, Guilhot F, Hjorth-Hansen H, Hughes TP, Kantarjian HM, Kim DW, Larson RA, Lipton JH, Mahon FX, Martinelli G, Mayer J, Muller MC, Niederwieser D, Pane F, Radich JP, Rousselot P, Saglio G, Saussele S, Schiffer C, Silver R, Simonsson B, Steegmann JL, Goldman JM, Hehlmann R. European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013. Blood. 2013;122:872–884. doi: 10.1182/blood-2013-05-501569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Pan Y, Lou J, Wang H, An N, Chen H, Zhang Q, Du X. CircBA9.3 supports the survival of leukaemic cells by up-regulating c-ABL1 or BCR-ABL1 protein levels. Blood Cells Mol Dis. 2018;73:38–44. doi: 10.1016/j.bcmd.2018.09.002. [DOI] [PubMed] [Google Scholar]
- 82.Ping L, Jian-Jun C, Chu-Shu L, Guang-Hua L, Ming Z. High circ_100053 predicts a poor outcome for chronic myeloid leukemia and is involved in imatinib resistance. Oncol Res. 2019 doi: 10.3727/096504018X15412701483326. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
- 83.Liu J, Kong F, Lou S, Yang D, Gu L. Global identification of circular RNAs in chronic myeloid leukemia reveals hsa_circ_0080145 regulates cell proliferation by sponging miR-29b. Biochem Biophys Res Commun. 2018;504:660–665. doi: 10.1016/j.bbrc.2018.08.154. [DOI] [PubMed] [Google Scholar]
- 84.Jabbour EJ, Faderl S, Kantarjian HM. Adult acute lymphoblastic leukemia. Mayo Clin Proc. 2005;80:1517–1527. doi: 10.4065/80.11.1517. [DOI] [PubMed] [Google Scholar]
- 85.Hunger SP, Lu X, Devidas M, Camitta BM, Gaynon PS, Winick NJ, Reaman GH, Carroll WL. Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: a report from the children’s oncology group. J. Clin. Oncol. 2012;30:1663–1669. doi: 10.1200/JCO.2011.37.8018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.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. doi: 10.2217/epi-2017-0142. [DOI] [PubMed] [Google Scholar]
- 87.Panda AC, Grammatikakis I, Kim KM, De S, Martindale JL, 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. doi: 10.1093/nar/gkw1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Chen J, Li Y, Zheng Q, Bao C, He J, Chen B, Lyu D, Zheng B, Xu Y, Long Z, Zhou Y, Zhu H, Wang Y, He X, Shi Y, Huang S. Circular RNA profile identifies circPVT1 as a proliferative factor and prognostic marker in gastric cancer. Cancer Lett. 2017;388:208–219. doi: 10.1016/j.canlet.2016.12.006. [DOI] [PubMed] [Google Scholar]
- 89.Rozman C, Montserrat E. Chronic lymphocytic leukemia. N Engl J Med. 1995;333:1052–1057. doi: 10.1056/NEJM199510193331606. [DOI] [PubMed] [Google Scholar]
- 90.Xia L, Wu L, Bao J, Li Q, Chen X, Xia H, Xia R. Circular RNA circ-CBFB promotes proliferation and inhibits apoptosis in chronic lymphocytic leukemia through regulating miR-607/FZD3/Wnt/beta-catenin pathway. Biochem Biophys Res Commun. 2018;503:385–390. doi: 10.1016/j.bbrc.2018.06.045. [DOI] [PubMed] [Google Scholar]
- 91.Wu W, Wu Z, Xia Y, Qin S, Li Y, Wu J, Liang J, Wang L, Zhu H, Fan L, Fu J, Xu W, Jin H, Li J. Downregulation of circ_0132266 in chronic lymphocytic leukemia promoted cell viability through miR-337-3p/PML axis. Aging (Albany NY) 2019;11:3561–3573. doi: 10.18632/aging.101997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Jiang M, Bennani NN, Feldman AL. Lymphoma classification update: T-cell lymphomas, Hodgkin lymphomas, and histiocytic/dendritic cell neoplasms. Expert Rev Hematol. 2017;10:239–249. doi: 10.1080/17474086.2017.1281122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Deng L, Liu G, Zheng C, Zhang L, Kang Y, Yang F. Circ-LAMP1 promotes T-cell lymphoblastic lymphoma progression via acting as a ceRNA for miR-615-5p to regulate DDR2 expression. Gene. 2019;701:146–151. doi: 10.1016/j.gene.2019.03.052. [DOI] [PubMed] [Google Scholar]
- 94.Dahl M, Daugaard I, Andersen MS, Hansen TB, Gronbaek K, Kjems J, Kristensen LS. Enzyme-free digital counting of endogenous circular RNA molecules in B-cell malignancies. Lab Invest. 2018;98:1657–1669. doi: 10.1038/s41374-018-0108-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Eisen MB, Spellman PT, Brown PO, Botstein D. Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci U S A. 1998;95:14863–14868. doi: 10.1073/pnas.95.25.14863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Rasche L, Kortum KM, Raab MS, Weinhold N. The impact of tumor heterogeneity on diagnostics and novel therapeutic strategies in multiple myeloma. Int J Mol Sci. 2019;20 doi: 10.3390/ijms20051248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Feng Y, Zhang L, Wu J, Khadka B, Fang Z, Gu J, Tang B, Xiao R, Pan G, Liu J. CircRNA circ_0000190 inhibits the progression of multiple myeloma through modulating miR-767-5p/MAPK4 pathway. J Exp Clin Cancer Res. 2019;38:54. doi: 10.1186/s13046-019-1071-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Glazar P, Papavasileiou P, Rajewsky N. circBase: a database for circular RNAs. RNA. 2014;20:1666–1670. doi: 10.1261/rna.043687.113. [DOI] [PMC free article] [PubMed] [Google Scholar]