Skip to main content

Long noncoding RNAs as regulators of pediatric acute myeloid leukemia

Abstract

Long noncoding RNAs (lncRNAs) are increasingly emerging as regulators across human development and disease, and many have been described in the context of hematopoiesis and leukemogenesis. These studies have yielded new molecular insights into the contribution of lncRNAs to AML development and revealed connections between lncRNA expression and clinical parameters in AML patients. In this mini review, we illustrate the versatile functions of lncRNAs in AML, with a focus on pediatric AML, and present examples that may serve as future therapeutic targets or predictive factors.

Background

Acute myeloid leukemia (AML) accounts for approximately 20% of acute leukemias in children [1]. Although the overall survival of children with AML has significantly increased as a result of intensified therapy, hematopoietic stem cell transplantation, and improved supportive care over the past decades, around 25% of all patients still cannot be cured [2] — highlighting the urgent need to transfer discoveries about the molecular features of pediatric AML into new therapeutic approaches. Among the recent scientific developments in this field, comprehensive studies have revealed that the molecular landscape of childhood AML is shaped not only by oncogenic mutations and cytogenetic alterations but also by global changes in DNA methylation and gene expression affecting both protein-coding genes and noncoding RNAs [3, 4]. Noncoding RNAs in particular are emerging as important regulators of hematopoiesis and leukemogenesis and represent a largely understudied space in the search for new therapeutic strategies.

Long noncoding RNAs (lncRNAs) — defined as transcripts longer than 200 nucleotides that lack open reading frames — represent the largest group of noncoding RNAs and constitute two-thirds of the human transcriptome [5]. Different structural domains enable their interaction with RNA, DNA, and proteins and thereby allow the regulation of every stage of gene expression. Apart from their versatile roles in gene regulation on every possible transcriptional and posttranscriptional level, lncRNAs can directly interact with signaling pathways and contribute to the function of organelles such as exosomes or mitochondria.

Molecular mechanisms and functions of lncRNAs

Based on the mechanistic interaction of lncRNAs with other molecules, four different archetypes of lncRNA functions — namely signal, decoy, guide, and scaffold — have been defined in a seminal work by Wang and Chang in 2011 [6]. As the first archetype, signal lncRNAs, which are under precise transcriptional control, act as a molecular signal reflecting a specific developmental stage, cellular background, or a response to stimuli [6,7,8,9]. LncRNAs belonging to the second archetype, decoy, bind and titrate away regulatory proteins or RNAs, thereby repressing transcription or translation of a target gene [10, 11]. Guide lncRNAs, which represent the third mechanistic archetype, direct regulatory protein complexes, chromatin modifiers, or transcription factors to their target site, resulting in either transcriptional activation or repression of the respective genomic locus [12, 13]. The fourth archetype, scaffold, describes lncRNAs as a structural platform at which different bound components of protein complexes or ribonucleoprotein complexes are assembled or can interact with each other [14, 15]. Even though increasing evidence now points toward complex lncRNA mechanisms that represent rather coexisting or overlapping features of the four classical archetypes, these four mechanistic subtypes still illustrate the wide range of possible modes of action of lncRNAs.

Nuclear-localized lncRNAs can exert independent regulatory effects on neighboring genes (function in cis) as well as on distant genes (function in trans), while cytoplasmic lncRNA mechanisms include competitive miRNA binding and interaction with translational proteins [16]. The most commonly demonstrated function of cis-acting nuclear lncRNAs, which are operating at their own site of transcription, is regulation of gene expression and chromatin modification [17]. The participation of cis-acting lncRNAs in transcriptional processes is supported by the high abundance of lncRNA genes in the proximity of regulatory elements of the human genome such as enhancers and promotors [18]. Protein-coding genes that are involved in transcriptional regulation, e.g., genes encoding transcription factors or chromatin modifiers, show a higher enrichment of closeby lncRNA genes than protein-coding genes of other functional categories, further indicating an essential contribution of cis-acting lncRNAs to the regulation of gene expression [19]. LncRNAs can act in cis to either activate or repress the expression of nearby genes through a variety of mechanisms. For instance, lncRNAs can activate gene expression in cis by recruiting proteins that establish spatial interactions such as chromatin loops, thereby enabling closer contact of an enhancer to the respective protein-coding gene [20, 21]. Other lncRNAs have been shown to activate gene expression of their target genes in cis in an transcript-independent manner by the process of their own transcription and splicing through recruiting cofactors, accumulation of transcriptional proteins, and establishing of activating chromatin marks [22, 23]. Conversely, cis-acting lncRNAs can also recruit chromatin modifiers that repress transcriptional activity at their genomic locus, such as the polycomb repressive complex 2 [24, 25]. Another lncRNA mechanism that results in decreased expression of the neighboring target gene is transcriptional interference: The transcription of a lncRNA can interfere with the transcription of the adjacent gene by impeding recruitment of necessary proteins such as transcription factors and chromatin remodeling proteins or by increasing nucleosome density, thereby preventing transcription factor access [26, 27]. For trans-acting lncRNAs, diverse functions in the modulation of distant gene expression have been demonstrated, with most of the studied examples exhibiting mechanisms that have been also described in the context of cis-acting lncRNAs. For instance, lncRNAs can facilitate transcriptional activation of distant target genes by the initiation of chromatin loop formation [28]. Similarly, several lncRNAs have been shown to repress transcription of target genes in trans through recruitment of chromatin-modifying complexes [7, 9, 29]. Another mechanism that has been described for cis-acting lncRNAs as well as for trans-acting lncRNAs is the formation of RNA-DNA hybrids, the so-called R-loops, that are recognized by transcription factors or chromatin modifiers and thereby lead to the activation or repression of transcription of the target gene [30].

Given their versatile cellular and molecular functions, it is no surprise that lncRNAs are involved in many essential physiological processes such as genomic imprinting and differentiation, as well as in the pathogenesis of diseases such as cancer, neurodegenerative disorders, and metabolomic diseases [31,32,33].

LncRNAs in hematopoiesis and AML

Hematopoietic differentiation is a tightly regulated, hierarchically ordered process coordinated by the expression of specific gene programs. Numerous lncRNAs have been characterized in the context of hematopoiesis including lncRNAs that are involved in hematopoietic fate decision and lncRNAs whose deregulation contributes to the malignant transformation of hematopoietic progenitor cells [34]. Recent studies identified unique stage- and lineage-specific lncRNA signatures in distinct blood cell populations indicating an important contribution of lncRNAs to the homeostasis and regulation of hematopoiesis [3, 35, 36]. Here, we review a selection of well-characterized lncRNAs that are involved at different levels of hematopoietic differentiation.

Fetal lncRNA H19 is one of the best-characterized lncRNAs in embryonic development and tumorigenesis. Physiologically downregulated after birth, H19 is expressed in almost every type of human cancer [37, 38]. During embryonic development, the lncRNA facilitates the transition from endothelial cells to hematopoietic stem cells (HSCs), whereas in adult hematopoiesis, it is essential for maintaining HSC quiescence, thereby regulating the long-term homeostasis of HSCs [39, 40]. In addition, H19 is overexpressed in AML and correlates with poor prognosis. In vitro knockdown of H19 leads to decreased proliferation and increased apoptosis in AML cell lines — further supporting its potential oncogenic effect in AML [41]. Another example of a lncRNA implicated in HSC homeostasis is LncHSC-2, a nuclear lncRNA, which is expressed in HSCs and hematopoietic progenitors [42]. LncHSC-2 regulates long-term self-renewal and lymphoid differentiation of HSCs by binding to Tcf3, a transcription factor that is essential for HSC proliferation and differentiation into myeloid-lymphoid progenitor cells [42].

In addition to these and other mechanistically studied examples of lncRNAs involved in HSC maintenance and maturation, several comprehensive transcriptomic studies identified hundreds of lncRNAs enriched in HSCs that are co-expressed with lineage-specific transcription factors, indicating that lncRNAs represent another important layer of the complex regulatory network that tunes hematopoietic differentiation [35, 36, 42]. Accordingly, lncRNAs are specifically enriched and functionally relevant not only in the context of HSCs but also in hematopoietic progenitor cell populations and mature blood cell populations. For instance, lncRNA HOTAIRM1 is highly expressed during granulocytic differentiation and contributes to the modulation of target genes in cis and in trans that are essential for proper myelopoiesis [43].

LINC00173 is another example of a lncRNA that is essentially involved in myeloid differentiation. We identified LINC00173 to be specifically expressed in mature granulocytes [3]. Upon knockdown in hematopoietic stem and progenitor cells (HSPCs), granulocytic differentiation and phagocytic capacity are impaired, whereas the erythroid lineage remained unaffected. Further analyses revealed a direct interaction between LINC00173 and PRC2, as well as differential H3K27 trimethylation at the promoter regions of genes involved in stemness, megakaryopoiesis, and erythropoiesis [3].

During erythroid differentiation, lncRNA EPS is enriched only in erythroid progenitor cells and promotes terminal erythrocytic differentiation by repressing pro-apoptotic pathways [44]. Within the lymphoid lineage, numerous lncRNAs that regulate differentiation and contribute to the immune response have been described. An example is lncRNA NeST, which is specifically expressed in CD4+ T-helper 1 cells and regulates the transcription of inflammatory genes through recruiting histone methyltransferase complexes [45, 46].

Dysregulation of the hematopoietic system results in uncontrolled proliferation of immature progenitor cells and in a block of proper differentiation, ultimately leading to the development of leukemia. Several lncRNAs have been shown to contribute to leukemogenesis [34]. In addition, lncRNAs may also serve as biomarkers or predictive factors in this disease [34]. It has been demonstrated that specific lncRNA expression profiles can be utilized to distinguish between different known molecular and cytogenetic AML subgroups and may serve as independent predictors of clinical outcome [47]. Using several examples, we illustrate different functions that have been described for individual lncRNAs in AML. We also briefly discuss lncRNA loci that may have functional consequences in AML cells independent from the encoded transcripts.

The lncRNA HOTAIR is highly expressed in AML and serves as a predictor for poor clinical outcome [48]. In vitro studies in primary AML blasts suggest an oncogenic function of HOTAIR, where it supposedly acts as a decoy for the tumor-suppressive microRNA miR-193a [48]. An alternative mode of action has also been described, where HOTAIR exerts its oncogenic effect in AML through EZH2-mediated epigenetic silencing of the tumor suppressor gene p15 [49].

LncRNA ANRIL, which is upregulated in both AML and ALL, acts as an oncogenic lncRNA by epigenetic silencing of its antisense tumor suppressor gene p15 [14, 50] As for other oncogenic lncRNAs, recent studies indicate a correlation of ANRIL expression to poor survival in patients with AML [51].

In contrast to the previous examples, lncRNA IRAIN is downregulated in AML cell lines and patients with high-risk AML, indicating that lncRNAs might not only act as oncogenes but as tumor suppressors in AML, too [52]. This lncRNA is transcribed antisense from the insulin-like growth factor type I receptor (IGF1R) locus, which is known to promote proliferation of AML cells through the PI3K/Akt signaling pathway [53, 54]. Mechanistically, IRAIN is involved in the formation of an intrachromosomal chromatin loop connecting the IGF1R promoter to a putative enhancer element [52]. However, the functional implications of this mechanism have yet to be elucidated. Clinical data further support the suggested tumor-suppressive function of IRAIN in AML, demonstrating a correlation between low IRAIN expression and poor prognosis in non-M3 acute myeloid leukemia patients [55].

While the majority of lncRNAs have yet to undergo in-depth characterization, this selection of examples provides a glimpse into the diversity of lncRNA functions in the context of hematopoiesis and AML. Even for these better-studied examples, it should be noted that mechanistic details remain elusive, due in part to the extensive experimental labor required to discern between RNA-dependent and -independent effects originating from lncRNA loci [56]. As a case in point, our group recently described MYNRL15 — a pan-myeloid leukemia dependency locus involved in genome topology, whose lncRNA product is dispensable for its dependency phenotype [57]. We found CTCF-enriched lncRNA loci (C-LNCs) like MYNRL15 to be enriched for leukemia vulnerabilities and provide a catalog (www.C-LNC.org) in hopes of facilitating the functional classification of lncRNAs and the discovery of new oncogenic vulnerabilities [57].

LncRNAs in pediatric AML

In contrast to many other malignant diseases, AML occurs in all age groups, but children account for only a small proportion of all patients with AML. The molecular landscape of pediatric AML differs significantly from the molecular profile of adult AML. Chromosomal aberrations are more common in children than in adults with AML [4, 58]. In addition, the genes that are frequently mutated in adult AML (NPM1, DNMT3A, IDH1, IDH2, RUNX1, TP53) are less often affected in children. Other genes, such as FLT3 or GATA2, differ in terms of the exact location and frequency of the mutations between pediatric and adult AML [4]. Given these biological and clinical differences, it is essential that we refrain from simply transferring new findings from adult cell lines, mouse models, and clinical cohorts of adult AML patients to the pediatric setting. Rather, investigations that focus specifically on pediatric AML are needed, to refine current risk stratification criteria and to develop novel therapeutic strategies for children with AML. While most current examples of lncRNAs with roles in AML have first been described in adult contexts, there is now an increasing number of studies characterizing lncRNAs in pediatric AML.

Our group described, for the first time, subtype-specific lncRNA signatures for six major cytogenetic subgroups of pediatric AML, namely, Down syndrome (DS) and non-DS acute megakaryoblastic leukemia (AMKL), inv[16], t[8;21], and AML with KMT2A rearrangement (t[9;11] and t[10;11]) [3]. In the transcriptional landscape of normal and malignant hematopoiesis, most DS- and non-DS-AMKL samples, and KMT2A-r samples, cluster in close proximity to HSCs. Their lncRNA expression profiles are characterized by the absence of myeloid expression programs. In contrast, all other pediatric AML samples clustered in proximity to normal myeloid progenitor cells. We further uncovered a core lncRNA stem cell signature that is shared between HSCs and AML blasts of all different pediatric AML subgroups. High expression of this core lncRNA stemness program is significantly correlated with poor survival in a cohort of adult AML patients [3].

Other studies have focused on the in-depth characterization of individual lncRNAs in pediatric AML. Here, we will summarize lncRNAs that have been implicated in pediatric AML (Table 1) and will exemplarily discuss several individual lncRNAs, for which molecular functions have been studied in the context of pediatric AML. Luo et al. found that the lncRNA HOTTIP is overexpressed in NPM1-mutated and KMT2A-r AML cases and predicts poor outcome [59]. Mechanistically, they showed that HOTTIP alters the three-dimensional structure of the nearby HOXA locus and binds to posterior HOXA sites as well as other genes critically involved in hematopoiesis and leukemogenesis, resulting in the activation of an AML-specific transcriptional program. Of note, HOTTIP expression is sufficient to initiate leukemic transformation of HSCs in mice. Knockout of HOTTIP perturbs leukemic proliferation and prolongs the survival in AML mouse models, suggesting a novel therapeutic option for the treatment of pediatric AML [59].

Table 1 LncRNAs implicated in pediatric acute myeloid leukemia

A recent elaborate study has identified lncRNA CDK6-AS1 as a novel regulator in pediatric AML [60]. In a pediatric patient cohort, CDK6-AS1 was significantly overexpressed and associated with higher minimal residual disease after induction therapy. High CDK6-AS1 levels contributed to an immature phenotype in healthy HSCs and primary AML blasts, whereas silencing of the lncRNA led to increased hematopoietic differentiation of HSCs and to a rescue of the pathogenic undifferentiated state of AML blasts. Mechanistically, the authors could show that CDK6-AS1 regulates expression of its neighboring gene CDK6 by sharing a bidirectional promotor, and that the common CDK6-AS1/CDK6 axis downregulates RUNX1 signaling, which is essential for early hematopoietic differentiation. In addition, CDK6-AS1 activates mitochondrial biogenesis in healthy HSCs as well as in pediatric AML blasts. Interestingly, mitochondrial targeting, using Tigecycline, sensitizes AML blasts with high CDK6-AS1 expression to chemotherapy, supporting the concept of a mitochondrial vulnerability in these blasts. Overall, these findings identified CDK6-AS1 as an important regulator of early hematopoietic differentiation and leukemogenesis of pediatric AML and uncovered therapeutics targeting mitochondrial biogenesis as a novel treatment strategy in pediatric AML [60].

UCA1 is an additional example of an oncogenic lncRNA in adult and pediatric AML. UCA1 has been shown to be upregulated by CEBPα-p30, the CEBPα isoform that results from CEBPA mutations recurrently found in AML patients [86]. In pediatric and adult AML cell lines, UCA1 is upregulated, and knockdown of the lncRNA impairs leukemic viability, migration, and invasion through binding of various microRNAs, such as miR-126, miR-204, miR96-5p, and miR296-3p [61,62,63,64]. Furthermore, UCA1 contributes to chemoresistance in pediatric AML by tethering miR-125a [65].

Other examples of relevant lncRNAs in childhood AML are MONC and MIR100HG, which are host genes for the homologous miRNA clusters miR-99a~125b-2 and miR-100~125b-1, respectively. These miRNA clusters are known to promote the progression of AMKL [87, 88]. Both lncRNA host genes are highly expressed in AMKL cells compared to cell lines of other pediatric AML subtypes. Lentiviral overexpression of MONC alters hematopoietic differentiation, independently of the expression of the oncogenic miRNA clusters [66].

LncRNA MEG3 is downregulated in adult and pediatric AML, supposedly by epigenetic modifications of its genomic locus. Mechanistically, MEG3 has been shown to activate p53 expression and DNMT3A, thereby inhibiting leukemogenesis [67]. Hypermethylation of the MEG3 promoter is associated with poor prognosis in adult AML patients [68]. In pediatric AML patients, higher expression of MEG3 correlates with better survival [69].

These are only a few examples of lncRNAs that have been characterized in the context of pediatric AML and that show correlation to clinically relevant subgroups and/or prognosis of the patients. In addition, a more general lncRNA scoring system based on the expression of 14 lncRNAs has been proposed to predict overall survival in children with AML [89].

HOXA10-AS: a novel oncogenic lncRNA in pediatric AML with KMT2A rearrangements

In a recently published study from our group, the antisense lncRNA HOXA10-AS was identified as an essential regulator of hematopoiesis and as a novel oncogenic lncRNA in the context of pediatric AML with KMT2A rearrangements (KMT2A-r AML) [70]. Along the genome, HOXA10-AS is located at the posterior end of the HOXA cluster — one of the four highly conserved HOX gene clusters. Tightly controlled spatiotemporal expression of the different HOX genes is crucial for hematopoietic differentiation, and dysregulation of HOX genes such as HOXA9 and HOXA7 by KMT2A fusion proteins is responsible for leukemic transformation in KMT2A-r AML [90]. The HOX gene clusters harbor numerous lncRNAs that are expressed in the same specific pattern as their protein-coding neighbors during differentiation, indicating their putative biological importance [9]. Previous studies revealed that HOX lncRNAs are capable of regulating the expression of neighboring or distant protein-coding HOX genes, as well as of independent effects on other signaling pathways [9, 12, 91]. Although a handful of HOX lncRNAs have undergone further characterization, the role of the vast majority of HOX lncRNAs in AML remains unknown.

HOXA10-AS is transcribed from the antisense strand relative to the protein-coding gene HOXA10 and microRNA mir-196b, both of which are involved in hematopoiesis and in the pathogenesis of KMT2A-r AML. In our study, we confirmed that HOXA10-AS is overexpressed in KMT2A-r AML as well (Fig. 1A). KMT2A rearrangements are the most frequent cytogenetic aberrations in pediatric AML and predominantly affect infants [58, 92]. Gain-of-function experiments in cell lines and primary blasts showed increased leukemic growth of KMT2A-r AML cells upon HOXA10-AS overexpression. In complementary loss-of-function assays using shRNA-mediated knockdown, CRISPR-Cas9-induced excision, and LNA-GapmeRs, we further demonstrated that the maintenance of KMT2A-r AML cells depends on high HOXA10-AS expression (Fig. 1B). During normal hematopoiesis, HOXA10-AS is specifically expressed in HSCs and strongly downregulated during hematopoietic differentiation, whereas the neighboring genes HOXA10 and mir-196b remain highly expressed in early myeloid progenitor cells (Fig. 1C). This strict stem cell-specific expression of HOXA10-AS suggests an independent regulatory circuit and cellular function separate from that of the nearby genes. Hematopoietic differentiation assays upon lentiviral overexpression of HOXA10-AS in HSPCs revealed impaired monocytic differentiation in HOXA10-AS overexpressing cells (Fig. 1D). The observations that ectopic expression of HOXA10-AS impairs monocytic differentiation and that HOXA10-AS is overexpressed in KMT2A-r AML are consistent with the fact that KMT2A-r AML predominantly manifests as a monoblastic leukemia (AML FAB M5) [58]. Regarding the mechanistic characterization of HOXA10-AS, we found that its effects in hematopoiesis and leukemogenesis were independent of changes in expression of the neighboring oncogenes, arguing against a regulatory role for HOXA10-AS on the HOXA cluster in cis. This was supported by subcellular localization studies, which showed that the HOXA10-AS is mainly located in the cytoplasm. Indeed, microarray-based gene expression analysis uncovered a possible trans mechanism for HOXA10-AS involving the upregulation of NF-κB target genes in HOXA10-AS expressing early monocytic progenitors and KMT2A-r AML (Fig. 1 A and D). Finally, we provided a proof of principle of how HOXA10-AS could be leveraged towards clinical implementation, by demonstrating HOXA10-AS as a prognostic marker in AML and potential therapeutic target in pediatric KMT2A-r AML [70].

Fig. 1
figure 1

HOXA10-AS, an example of a lncRNA regulator of hematopoiesis and pediatric leukemia. A HOXA10-AS, is overexpressed in pediatric AML with KMT2A rearrangements, where it increases leukemic proliferation via activation of the NF-κB signaling pathway. B Knockdown of HOXA10-AS using shRNAs leads to reduced growth of KMT2A-r AML patient blasts in vivo. C HOX10-AS is specifically expressed in hematopoietic stem cells and downregulated during hematopoietic differentiation. D Upon overexpression in hematopoietic stem and progenitor cells (HSPCs), HOXA10-AS impairs monocytic differentiation through the activation of NF-κB target genes

Conclusion

LncRNAs are emerging as regulators of hematopoiesis and AML pathogenesis, and knowledge about their individual effects is rapidly increasing. However, extensive functional research is required before we gain a complete understanding of the complex regulatory networks surrounding lncRNAs and their interplay with known oncogenic drivers. While individual examples continue to provide valuable information about the roles of lncRNAs and how they might serve as novel therapeutic targets or prognostic factors in AML, research on lncRNAs in pediatric AML still lags behind adult AML. Thus, investigations of lncRNAs such as HOXA10-AS add important insights on the regulatory roles of lncRNAs in general, as well as crucial knowledge about the specific pathogenesis of pediatric AML, both of which will hopefully contribute to a comprehensive view and new therapies for this disease.

Availability of data and materials

Not applicable.

Abbreviations

AML:

Acute myeloid leukemia

AMKL:

Acute megakaryoblastic leukemia

DS:

Down syndrome

HSC:

Hematopoietic stem cell

HSPC:

Hematopoietic stem and progenitor cell

KMT2A-r AML:

Acute myeloid leukemia with KMT2A rearrangements

lncRNA:

Long noncoding RNA

shRNA:

Short hairpin RNA

References

  1. Erdmann F, Kaatsch P, Grabow D, Spix C (2020) German Childhood Cancer Registry - Annual Report 2019 (1980-2018)

    Google Scholar 

  2. Rasche M, Zimmermann M, Borschel L, Bourquin J-P, Dworzak M, Klingebiel T et al (2018) Successes and challenges in the treatment of pediatric acute myeloid leukemia: a retrospective analysis of the AML-BFM trials from 1987 to 2012. Leukemia 32(10):2167–2177

    Article  PubMed  PubMed Central  Google Scholar 

  3. Schwarzer A, Emmrich S, Schmidt F, Beck D, Ng M, Reimer C et al (2017) The non-coding RNA landscape of human hematopoiesis and leukemia. Nat Commun 8(1):218

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Bolouri H, Farrar JE, Triche TJ, Ries RE, Lim EL, Alonzo TA et al (2018) The molecular landscape of pediatric acute myeloid leukemia reveals recurrent structural alterations and age-specific mutational interactions. Nat Med 24(1):103–112

    Article  CAS  PubMed  Google Scholar 

  5. Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D et al (2009) Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature 458(7235):223–227

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Wang KC, Chang HY (2011) Molecular mechanisms of long noncoding RNAs. Mol Cell 43(6):904–914

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Huarte M, Guttman M, Feldser D, Garber M, Koziol MJ, Kenzelmann-Broz D et al (2010) A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell 142(3):409–419

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Loewer S, Cabili MN, Guttman M, Loh Y-H, Thomas K, Park IH et al (2010) Large intergenic non-coding RNA-RoR modulates reprogramming of human induced pluripotent stem cells. Nat Genet 42(12):1113–1117

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA et al (2007) Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell 129(7):1311–1323

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ, Pandolfi PP (2010) A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature 465(7301):1033–1038

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Hung T, Wang Y, Lin MF, Koegel AK, Kotake Y, Grant GD et al (2011) Extensive and coordinated transcription of noncoding RNAs within cell-cycle promoters. Nat Genet 43(7):621–629

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wang KC, Yang YW, Liu B, Sanyal A, Corces-Zimmerman R, Chen Y et al (2011) A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature 472(7341):120–124

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ et al (2010) Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 464(7291):1071–1076

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kotake Y, Nakagawa T, Kitagawa K, Suzuki S, Liu N, Kitagawa M et al (2011) Long non-coding RNA ANRIL is required for the PRC2 recruitment to and silencing of p15(INK4B) tumor suppressor gene. Oncogene 30(16):1956–1962

    Article  CAS  PubMed  Google Scholar 

  15. Ly H, Blackburn EH, Parslow TG (2003) Comprehensive structure-function analysis of the core domain of human telomerase RNA. Mol Cell Biol 23(19):6849–6856

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Guttman M, Rinn JL (2012) Modular regulatory principles of large non-coding RNAs. Nature 482(7385):339–346

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gil N, Ulitsky I (2020) Regulation of gene expression by cis-acting long non-coding RNAs. Nat Rev Genet 21(2):102–117

    Article  CAS  PubMed  Google Scholar 

  18. Marques AC, Hughes J, Graham B, Kowalczyk MS, Higgs DR, Ponting CP (2013) Chromatin signatures at transcriptional start sites separate two equally populated yet distinct classes of intergenic long noncoding RNAs. Genome Biol 14(11):R131

    Article  PubMed  PubMed Central  Google Scholar 

  19. Ponjavic J, Oliver PL, Lunter G, Ponting CP (2009) Genomic and transcriptional co-localization of protein-coding and long non-coding RNA pairs in the developing brain. PLoS Genet 5(8):e1000617

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Xiang J-F, Yin Q-F, Chen T, Zhang Y, Zhang X-O, Wu Z et al (2014) Human colorectal cancer-specific CCAT1-L lncRNA regulates long-range chromatin interactions at the MYC locus. Cell Res 24(5):513–531

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lai F, Orom UA, Cesaroni M, Beringer M, Taatjes DJ, Blobel GA et al (2013) Activating RNAs associate with mediator to enhance chromatin architecture and transcription. Nature 494(7438):497–501

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Engreitz JM, Haines JE, Perez EM, Munson G, Chen J, Kane M et al (2016) Local regulation of gene expression by lncRNA promoters, transcription and splicing. Nature 539(7629):452–455

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Gil N, Ulitsky I (2018) Production of spliced long noncoding RNAs specifies regions with increased enhancer activity. Cell Syst 7(5):537–547.e3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Zhao J, Sun BK, Erwin JA, Song J-J, Lee JT (2008) Polycomb proteins targeted by a short repeat RNA to the mouse X chromosome. Science 322(5902):750–756

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Pandey RR, Mondal T, Mohammad F, Enroth S, Redrup L, Komorowski J et al (2008) Kcnq1ot1 antisense noncoding RNA mediates lineage-specific transcriptional silencing through chromatin-level regulation. Mol Cell 32(2):232–246

    Article  CAS  PubMed  Google Scholar 

  26. Bumgarner SL, Dowell RD, Grisafi P, Gifford DK, Fink GR (2009) Toggle involving cis-interfering noncoding RNAs controls variegated gene expression in yeast. Proc Natl Acad Sci U S A 106(43):18321–18326

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Hainer SJ, Pruneski JA, Mitchell RD, Monteverde RM, Martens JA (2011) Intergenic transcription causes repression by directing nucleosome assembly. Genes Dev 25(1):29–40

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Grossi E, Raimondi I, Goñi E, González J, Marchese FP, Chapaprieta V et al (2020) A lncRNA-SWI/SNF complex crosstalk controls transcriptional activation at specific promoter regions. Nat Commun 11(1):936

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Holdt LM, Hoffmann S, Sass K, Langenberger D, Scholz M, Krohn K et al (2013) Alu elements in ANRIL non-coding RNA at chromosome 9p21 modulate atherogenic cell functions through trans-regulation of gene networks. PLoS Genet 9(7):e1003588

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ariel F, Lucero L, Christ A, Mammarella MF, Jegu T, Veluchamy A et al (2020) R-Loop Mediated trans action of the APOLO long noncoding RNA. Mol Cell 77(5):1055–1065.e4

    Article  CAS  PubMed  Google Scholar 

  31. Huarte M (2015) The emerging role of lncRNAs in cancer. Nat Med 21(11):1253–1261

    Article  CAS  PubMed  Google Scholar 

  32. Morán I, Akerman I, van de Bunt M, Xie R, Benazra M, Nammo T et al (2012) Human β cell transcriptome analysis uncovers lncRNAs that are tissue-specific, dynamically regulated, and abnormally expressed in type 2 diabetes. Cell Metab 16(4):435–448

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Zhou S, Yu X, Wang M, Meng Y, Song D, Yang H et al (2021) Long non-coding RNAs in pathogenesis of neurodegenerative diseases. Front Cell Dev Biol 9:719247

    Article  PubMed  PubMed Central  Google Scholar 

  34. Ng M, Heckl D, Klusmann J-H (2019) The regulatory roles of long noncoding RNAs in acute myeloid leukemia. Front Oncol 9:570

    Article  PubMed  PubMed Central  Google Scholar 

  35. Wu Z, Gao S, Zhao X, Chen J, Keyvanfar K, Feng X et al (2019) Long noncoding RNAs of single hematopoietic stem and progenitor cells in healthy and dysplastic human bone marrow. Haematologica 104(5):894–906

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Cabezas-Wallscheid N, Klimmeck D, Hansson J, Lipka DB, Reyes A, Wang Q et al (2014) Identification of regulatory networks in HSCs and their immediate progeny via integrated proteome, transcriptome, and DNA methylome analysis. Cell Stem Cell 15(4):507–522

    Article  CAS  PubMed  Google Scholar 

  37. Raveh E, Matouk IJ, Gilon M, Hochberg A (2015) The H19 long non-coding RNA in cancer initiation, progression and metastasis - a proposed unifying theory. Mol Cancer 14:184

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Gabory A, Jammes H, Dandolo L (2010) The H19 locus: role of an imprinted non-coding RNA in growth and development. Bioessays 32(6):473–480

    Article  CAS  PubMed  Google Scholar 

  39. Venkatraman A, He XC, Thorvaldsen JL, Sugimura R, Perry JM, Tao F et al (2013) Maternal imprinting at the H19-Igf2 locus maintains adult haematopoietic stem cell quiescence. Nature 500(7462):345–349

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Zhou J, Xu J, Zhang L, Liu S, Ma Y, Wen X et al (2019) Combined single-cell profiling of lncRNAs and functional screening reveals that H19 is pivotal for embryonic hematopoietic stem cell development. Cell Stem Cell 24(2):285–298.e5

    Article  CAS  PubMed  Google Scholar 

  41. Zhang T-J, Zhou J-D, Zhang W, Lin J, Ma J-C, Wen X-M et al (2018) H19 overexpression promotes leukemogenesis and predicts unfavorable prognosis in acute myeloid leukemia. Clin Epigenetics 10:47

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Luo M, Jeong M, Sun D, Park HJ, Rodriguez BAT, Xia Z et al (2015) Long non-coding RNAs control hematopoietic stem cell function. Cell Stem Cell 16(4):426–438

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Zhang X, Lian Z, Padden C, Gerstein MB, Rozowsky J, Snyder M et al (2009) A myelopoiesis-associated regulatory intergenic noncoding RNA transcript within the human HOXA cluster. Blood 113(11):2526–2534

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Hu W, Yuan B, Flygare J, Lodish HF (2011) Long noncoding RNA-mediated anti-apoptotic activity in murine erythroid terminal differentiation. Genes Dev 25(24):2573–2578

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Gomez JA, Wapinski OL, Yang YW, Bureau J-F, Gopinath S, Monack DM et al (2013) The NeST long ncRNA controls microbial susceptibility and epigenetic activation of the interferon-γ locus. Cell 152(4):743–754

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Collier SP, Collins PL, Williams CL, Boothby MR, Aune TM (2012) Cutting edge: influence of Tmevpg1, a long intergenic noncoding RNA, on the expression of Ifng by Th1 cells. J Immunol 189(5):2084–2088

    Article  CAS  PubMed  Google Scholar 

  47. Beck D, Thoms JAI, Palu C, Herold T, Shah A, Olivier J et al (2018) A four-gene LincRNA expression signature predicts risk in multiple cohorts of acute myeloid leukemia patients. Leukemia 32(2):263–272

    Article  CAS  PubMed  Google Scholar 

  48. Xing C, Hu X, Xie F, Yu Z, Li H, Zhou B et al (2015) Long non-coding RNA HOTAIR modulates c-KIT expression through sponging miR-193a in acute myeloid leukemia. FEBS Lett 589(15):1981–1987

    Article  CAS  PubMed  Google Scholar 

  49. Gao S, Zhou B, Li H, Huang X, Wu Y, Xing C et al (2018) Long noncoding RNA HOTAIR promotes the self-renewal of leukemia stem cells through epigenetic silencing of p15. Exp Hematol 67:32–40.e3

    Article  CAS  PubMed  Google Scholar 

  50. Yu W, Gius D, Onyango P, Muldoon-Jacobs K, Karp J, Feinberg AP et al (2008) Epigenetic silencing of tumour suppressor gene p15 by its antisense RNA. Nature 451(7175):202–206

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Tan Z, Zhu K, Yin Y, Luo Z (2021) Long non-coding RNA ANRIL is a potential indicator of disease progression and poor prognosis in acute myeloid leukemia. Mol Med Rep 23(2):112.

  52. Sun J, Li W, Sun Y, Yu D, Wen X, Wang H et al (2014) A novel antisense long noncoding RNA within the IGF1R gene locus is imprinted in hematopoietic malignancies. Nucleic Acids Res 42(15):9588–9601

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Chapuis N, Tamburini J, Cornillet-Lefebvre P, Gillot L, Bardet V, Willems L et al (2010) Autocrine IGF-1/IGF-1R signaling is responsible for constitutive PI3K/Akt activation in acute myeloid leukemia: therapeutic value of neutralizing anti-IGF-1R antibody. Haematologica 95(3):415–423

    Article  CAS  PubMed  Google Scholar 

  54. Xu Q, Simpson S-E, Scialla TJ, Bagg A, Carroll M (2003) Survival of acute myeloid leukemia cells requires PI3 kinase activation. Blood 102(3):972–980

    Article  CAS  PubMed  Google Scholar 

  55. Pashaiefar H, Izadifard M, Yaghmaie M, Montazeri M, Gheisari E, Ahmadvand M et al (2018) Low expression of long noncoding RNA IRAIN is associated with poor prognosis in non-M3 acute myeloid leukemia patients. Genet Test Mol Biomarkers 22(5):288–294

    Article  CAS  PubMed  Google Scholar 

  56. Kopp F, Mendell JT (2018) Functional classification and experimental dissection of long noncoding RNAs. Cell 172(3):393–407

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ng M, Verboon L, Issa H et al (2021) Myeloid leukemia vulnerabilities at CTCF-enriched long noncoding RNA loci, PREPRINT (Version 1) available at Research Square

  58. von Neuhoff C, Reinhardt D, Sander A, Zimmermann M, Bradtke J, Betts DR et al (2010) Prognostic impact of specific chromosomal aberrations in a large group of pediatric patients with acute myeloid leukemia treated uniformly according to trial AML-BFM 98. J Clin Oncol Off J Am Soc Clin Oncol 28(16):2682–2689

    Article  CAS  Google Scholar 

  59. Luo H, Zhu G, Xu J, Lai Q, Yan B, Guo Y et al (2019) HOTTIP lncRNA promotes hematopoietic stem cell self-renewal leading to AML-like disease in mice. Cancer Cell 36(6):645–659.e8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Porcù E, Benetton M, Bisio V, Da Ros A, Tregnago C, Borella G et al (2021) The long non-coding RNA CDK6-AS1 overexpression impacts on acute myeloid leukemia differentiation and mitochondrial dynamics. iScience 24(11):103350

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Sun M-D, Zheng Y-Q, Wang L-P, Zhao H-T, Yang S (2018) Long noncoding RNA UCA1 promotes cell proliferation, migration and invasion of human leukemia cells via sponging miR-126. Eur Rev Med Pharmacol Sci 22(8):2233–2245

    PubMed  Google Scholar 

  62. Liang Y, Li E, Zhang H, Zhang L, Tang Y, Wanyan Y (2020) Silencing of lncRNA UCA1 curbs proliferation and accelerates apoptosis by repressing SIRT1 signals by targeting miR-204 in pediatric AML. J Biochem Mol Toxicol 34(3):e22435

    Article  CAS  PubMed  Google Scholar 

  63. Li JJ, Chen XF, Wang M, Zhang PP, Zhang F, Zhang JJ (2020) Long non-coding RNA UCA1 promotes autophagy by targeting miR-96-5p in acute myeloid leukaemia. Clin Exp Pharmacol Physiol 47(5):877–885

    Article  CAS  PubMed  Google Scholar 

  64. Li J, Wang M, Chen X (2020) Long non-coding RNA UCA1 modulates cell proliferation and apoptosis by regulating miR-296-3p/Myc axis in acute myeloid leukemia. Cell Cycle 19(12):1454–1465

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Zhang Y, Liu Y, Xu X (2018) Knockdown of LncRNA-UCA1 suppresses chemoresistance of pediatric AML by inhibiting glycolysis through the microRNA-125a/hexokinase 2 pathway. J Cell Biochem 119(7):6296–6308

    Article  CAS  PubMed  Google Scholar 

  66. Emmrich S, Streltsov A, Schmidt F, Thangapandi VR, Reinhardt D, Klusmann J-H (2014) LincRNAs MONC and MIR100HG act as oncogenes in acute megakaryoblastic leukemia. Mol Cancer 13:171

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Lyu Y, Lou J, Yang Y, Feng J, Hao Y, Huang S et al (2017) Dysfunction of the WT1-MEG3 signaling promotes AML leukemogenesis via p53-dependent and -independent pathways. Leukemia 31(12):2543–2551

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Benetatos L, Hatzimichael E, Dasoula A, Dranitsaris G, Tsiara S, Syrrou M et al (2010) CpG methylation analysis of the MEG3 and SNRPN imprinted genes in acute myeloid leukemia and myelodysplastic syndromes. Leuk Res 34(2):148–153

    Article  CAS  PubMed  Google Scholar 

  69. Xue H, Gao H, Xia H, Li S, Li N, Duan Y et al (2021) Prognostic significance of long non coding maternally expressed gene 3 in pediatric acute myeloid leukemia. Medicine (Baltimore) 100(35):e26959

    Article  CAS  Google Scholar 

  70. Al-Kershi S, Bhayadia R, Ng M, Verboon L, Emmrich S, Gack L et al (2019) The stem cell-specific long noncoding RNA HOXA10-AS in the pathogenesis of KMT2A-rearranged leukemia. Blood Adv 3(24):4252–4263

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Fang X, Pan X, Mai H, Yuan X, Liu S, Wen F (2021) LINC00998 functions as a novel tumor suppressor in acute myeloid leukemia via regulating the ZFP36 ring finger protein/mammalian target of rapamycin complex 2 axis. Bioengineered 12(2):10363–10372

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Connerty P, Moles E, de Bock CE, Jayatilleke N, Smith JL, Meshinchi S et al (2021) Development of siRNA-loaded lipid nanoparticles targeting long non-coding RNA LINC01257 as a novel and safe therapeutic approach for t(8;21) pediatric acute myeloid leukemia. Pharmaceutics 13(10):1681

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Xue H, Gao H, Xia H, Li S, Li N, Gao C et al (2021) IncRNA MVIH correlates with disease features, predicts treatment response and survival in pediatric acute myeloid leukemia. J Clin Lab Anal 35(4):e23739

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Lei W, Lin J, Liu F, Chen N (2021) Long noncoding RNA GAS6 antisense RNA1 silencing attenuates the tumorigenesis of acute myeloid leukemia cells through targeting microRNA-370-3p/Tetraspanin3 axis. Clin Hemorheol Microcirc 78(1):69–81

    Article  CAS  PubMed  Google Scholar 

  75. Sheng H, Zhang J, Ma Y, Zhang Y, Dai Y, Jiang R (2021) lncRNA FBXL19-AS1 is a diagnosis biomarker for paediatric patients with acute myeloid leukemia. J Gene Med 23(3):e3317

    Article  CAS  PubMed  Google Scholar 

  76. Wang X, Li W, Chen Y, Zhou L (2021) Long non-coding RNA SNHG14 affects the proliferation and apoptosis of childhood acute myeloid leukaemia cells by modulating the miR-193b-3p/MCL1 axis. Mol Med Rep 23(2):90

    Article  CAS  PubMed  Google Scholar 

  77. Dou B, Jiang Z, Chen X, Wang C, Wu J, An J et al (2020) Oncogenic long noncoding RNA DARS-AS1 in childhood acute myeloid leukemia by binding to microRNA-425. Technol Cancer Res Treat 19:1533033820965580

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Zhang X, Yang L, Xu G (2020) Silencing of long noncoding RNA TUG1 inhibits viability and promotes apoptosis of acute myeloid leukemia cells by targeting microRNA-221-3p/KIT axis. Clin Hemorheol Microcirc 76(3):425–437

    Article  CAS  PubMed  Google Scholar 

  79. Ma L, Wang Y-Y, Jiang P (2020) LncRNA LINC00909 promotes cell proliferation and metastasis in pediatric acute myeloid leukemia via miR-625-mediated modulation of Wnt/β-catenin signaling. Biochem Biophys Res Commun 527(3):654–661

    Article  CAS  PubMed  Google Scholar 

  80. Wang W-T, Chen T-Q, Zeng Z-C, Pan Q, Huang W, Han C et al (2020) The lncRNA LAMP5-AS1 drives leukemia cell stemness by directly modulating DOT1L methyltransferase activity in MLL leukemia. J Hematol Oncol 13(1):78

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Wang J, Liu Z-H, Yu L-J (2019) Long non-coding RNA LINC00641 promotes cell growth and migration through modulating miR-378a/ZBTB20 axis in acute myeloid leukemia. Eur Rev Med Pharmacol Sci 23(17):7498–7509

    CAS  PubMed  Google Scholar 

  82. Guan X, Wen X, Xiao J, An X, Yu J, Guo Y (2019) Lnc-SOX6-1 upregulation correlates with poor risk stratification and worse treatment outcomes, and promotes cell proliferation while inhibits apoptosis in pediatric acute myeloid leukemia. Int J Lab Hematol 41(2):234–241

    Article  PubMed  Google Scholar 

  83. El-Khazragy N, Elayat W, Matbouly S, Seliman S, Sami A, Safwat G et al (2019) The prognostic significance of the long non-coding RNAs “CCAT1, PVT1” in t(8;21) associated acute myeloid leukemia. Gene 707:172–177

    Article  CAS  PubMed  Google Scholar 

  84. Fernando TR, Contreras JR, Zampini M, Rodriguez-Malave NI, Alberti MO, Anguiano J et al (2017) The lncRNA CASC15 regulates SOX4 expression in RUNX1-rearranged acute leukemia. Mol Cancer 16(1):126

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Morenos L, Chatterton Z, Ng JL, Halemba MS, Parkinson-Bates M, Mechinaud F et al (2014) Hypermethylation and down-regulation of DLEU2 in paediatric acute myeloid leukaemia independent of embedded tumour suppressor miR-15a/16-1. Mol Cancer 13:123

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Hughes JM, Legnini I, Salvatori B, Masciarelli S, Marchioni M, Fazi F et al (2015) C/EBPα-p30 protein induces expression of the oncogenic long non-coding RNA UCA1 in acute myeloid leukemia. Oncotarget 6(21):18534–18544

    Article  PubMed  PubMed Central  Google Scholar 

  87. Emmrich S, Rasche M, Schöning J, Reimer C, Keihani S, Maroz A et al (2014) miR-99a/100~125b tricistrons regulate hematopoietic stem and progenitor cell homeostasis by shifting the balance between TGFβ and Wnt signaling. Genes Dev 28(8):858–874

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Alejo-Valle O, Weigert K, Bhayadia R, Ng M, Issa H et al (2022) The megakaryocytic transcription factor ARID3A suppresses leukemia pathogenesis. Blood 139(5):651–665

  89. Guo S, Li B, Xu X, Wang W, Wang S, Lv T et al (2020) Construction of a 14-lncRNA risk score system predicting survival of children with acute myelocytic leukemia. Exp Ther Med 20(2):1521–1531

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Ayton PM, Cleary ML (2003) Transformation of myeloid progenitors by MLL oncoproteins is dependent on Hoxa7 and Hoxa9. Genes Dev 17(18):2298–2307

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Zhao H, Zhang X, Frazão JB, Condino-Neto A, Newburger PE (2013) HOX antisense lincRNA HOXA-AS2 is an apoptosis repressor in all trans retinoic acid treated NB4 promyelocytic leukemia cells. J Cell Biochem 114(10):2375–2383

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Pui CH, Raimondi SC, Srivastava DK, Tong X, Behm FG, Razzouk B et al (2000) Prognostic factors in infants with acute myeloid leukemia. Leukemia 14(4):684–687

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

S. Neyazi was a recipient of Mildred-Scheel doctoral research funding from the German Cancer Aid. This work was supported by funding to J. H. K from the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation program (grant agreement #714226). J. H. K. is a recipient of the St. Baldrick’s Robert J. Arceci Innovation Award.

Author information

Authors and Affiliations

Authors

Contributions

SN wrote the manuscript. MN, DH, and JHK edited the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Sina Neyazi.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

J. H. K. has advisory roles for bluebird bio, Novartis, Roche, and Jazz Pharmaceuticals. The authors disclose that their published work is highlighted in this mini review.

Additional information

Publisher’s Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Neyazi, S., Ng, M., Heckl, D. et al. Long noncoding RNAs as regulators of pediatric acute myeloid leukemia. Mol Cell Pediatr 9, 10 (2022). https://doi.org/10.1186/s40348-022-00142-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40348-022-00142-2