Expression of microRNAs, a new class of noncoding RNAs that hybridize to target messenger RNA and regulate their translation into proteins, has been recently demonstrated to be altered in acute myeloid leukemia (AML). Distinctive patterns of increased expression and/or silencing of multiple microRNAs (microRNA signatures) have been associated with specific cytogenetic and molecular subsets of AML. Changes in the expression of several microRNAs altered in AML have been shown to have functional relevance in leukemogenesis, with some microRNAs acting as oncogenes and others as tumor suppressors. Both microRNA signatures and a single microRNA (ie, miR-181a) have been shown to supply prognostic information complementing that gained from cytogenetics, gene mutations, and altered gene expression. Moreover, it has been demonstrated experimentally that antileukemic effects can be achieved by modulating microRNA expression by pharmacologic agents and/or increasing low endogenous levels of microRNAs with tumor suppressor function by synthetic microRNA oligonucleotides, or down-regulating high endogenous levels of leukemogenic microRNAs by antisense oligonucleotides (antagomirs). Therefore, it is reasonable to predict the development of novel microRNA-based therapeutic approaches in AML. We review herein results of current studies analyzing changes of microRNA expression in AML and discuss their potential biologic, diagnostic, and prognostic relevance.

MicroRNAs are naturally occurring 18- to 25-nucleotide RNAs that are cleaved from 70- to 100-nucleotide hairpin precursors by a complex protein system that includes the RNase III Drosha and Dicer, members of the Argonaute family.1,2  Mature microRNAs typically hybridize to the 3′-untranslated regions of protein-coding messenger RNAs (mRNAs) and cause their post-transcriptional repression and/or degradation in proliferating cells,1,2  although emerging data indicate that microRNAs can also regulate gene expression through binding to “seedless” 3′-untranslated region microRNA recognition elements3  or increase translation of target mRNAs on cell cycle arrest.4  MicroRNAs, whose nomenclature is described by Griffiths-Jones et al,5  regulate normal cell homeostasis and are involved in many physiologic processes, including hematopoiesis.6-8  Recently, dysregulation of microRNAs has been shown in many types of solid tumors and leukemia.9  Direct involvement of microRNAs in cancer has been suggested by a study demonstrating that several microRNAs are localized in genomic regions associated with cancer, such as breakpoint regions in chromosome aberrations involving oncogenes or tumor suppressor genes, minimal regions of loss of heterozygosity, minimal regions of amplification, and at loci close to fragile sites and integration sites of the human papilloma virus.10  Supporting their role in carcinogenesis, microRNAs mapped to the chromosomal regions deleted in cancer samples were found expressed at low levels,9  and several functional studies confirmed the important role of microRNA deregulation in hematologic malignancies, including acute myeloid leukemia (AML).9,11-19 

Profiling and functional experiments revealed that several microRNAs play a role in the regulation of hematopoiesis.6-8  Hence, it was not surprising that many microRNAs are differentially expressed between AML blasts and normal cells.20-23  However, microRNA signatures derived by comparing microRNA expression between normal and malignant cells have shared few similarities among different studies. This lack of uniformity could be explained by the use of normal CD34+ cells obtained under different conditions, such as after growth factor mobilization versus collected directly from the bone marrow (BM) with no mobilization. It is possible that the use of growth factors could change microRNA expression in CD34+ cells because similar changes have been observed for mRNA expression.24  In addition, some studies used unselected BM mononuclear cells from healthy donors instead of CD34+ cells as controls.20  The use of different platforms for microRNA profiling and differences in the frequency of cytogenetic and/or molecular genetic alterations, shown to affect microRNA expression (see “Correlations of microRNA expression with cytogenetics in AML” and “Correlations of microRNA expression with molecular markers in CN-AML”), within AML patient cohorts analyzed could also explain discordant results.20-23  Although a comparison of microRNA expression between normal CD34+ cells and leukemic stem cells instead of AML blasts could potentially be more informative, to our knowledge, no such study has yet been reported.

Similar to a pioneering study, which demonstrated that gene-expression profiling (GEP) can distinguish between AML and acute lymphoblastic leukemia (ALL),25  changes in microRNA expression were also shown to define lineage-specific leukemia. Using a bead-based microRNA-expression profiling assay, Mi et al20  separated patients with AML from those with ALL based on 27 differentially expressed microRNAs (21 up-regulated and 6 down-regulated in AML compared with ALL), 4 of which, the up-regulated let-7b and miR-223, and down-regulated miR-128a and miR-128b, were the most discriminatory.

Cytogenetically, AML is a very heterogeneous disease, with approximately 200 chromosomal abnormalities identified as recurrent.26-28  Whereas many of these aberrations are very rare, being detected in a few patients worldwide, others occur more frequently and have been associated with response to treatment and survival.29,30  Among the more frequent abnormalities, t(8;21)(q22;q22) and inv(16)(p13q22) [and its variant t(16;16)(p13;q22)], whose presence characterizes patients with core-binding factor AML (CBF-AML), and t(15;17)(q22;q12-q21) or its variants, a translocation pathognomonic for acute promyelocytic leukemia (APL), bestow a relatively favorable outcome. In contrast, patients with balanced translocations involving band 11q23 and the MLL gene [t(11q23)/MLL] other than t(9;11)(p22;q23), inv(3)(q21q26) or t(3;3)(q21;q26), t(6;9)(p23;q34), deletion or loss of 5q, monosomy 7, structural abnormalities of 17p or a complex karyotype [defined as more than or equal to 3 chromosome aberrations in the absence of t(15;17), t(8;21), inv(16) or t(16;16), t(9;11), t(11q23), t(6;9), and inv(3) or t(3;3)31 ] have a very poor prognosis.26,27,29-34  Patients with other chromosome aberrations or those with cytogenetically normal AML (CN-AML) have better outcome, but not as good as patients with CBF-AML and APL, and are classified as having an intermediate cytogenetic risk.26,27,29-33,35 

Recent studies have shown using microRNA-expression profiling that several of the aforementioned cytogenetic subtypes of AML are associated with microRNA signatures. Each of the favorable cytogenetic-risk subtypes, namely, CBF-AML with t(8;21), CBF-AML with inv(16) or t(16;16), and APL with t(15;17), has had a unique microRNA-expression profile that distinguished them from each other and from other AML subtypes.21,23,36,37  Dixon-McIver et al37  identified a characteristic signature in APL patients with t(15;17), which comprised 7 up-regulated microRNAs transcribed from genes located in the imprinted domain at chromosome band 14q32 (miR-127, miR-154, miR-154*, miR-299, miR-323, miR-368, and miR-370), and 9 down-regulated microRNAs (miR-17-3p, miR-185, miR-187, miR-194, miR-200a, miR-200b, miR-200c, miR-330, and miR-339). In another study,21  a signature from a cluster that consisted of samples from t(15;17)-positive APL patients displayed a strong up-regulation of miR-127, miR-134, miR-299-5p, miR-323, miR-376a, and miR-382. In a third study,36  3 of the 7 microRNAs identified as class-discriminatory, miR-224, miR-368, and miR-382, were almost exclusively overexpressed in the t(15;17) samples, whereas miR-17-5p, miR-20a, miR-126, and miR-126* were down-regulated.

In contrast, overexpression of miR-126 and miR-126* occurred in CBF-AML samples, both those with t(8;21)21,23,36  and those with inv(16).36  A colony-forming and replating assay revealed cooperation between miR-126 and t(8;21)/RUNX1-RUNXT1(AML1/ETO) in enhancing proliferation of mouse BM progenitor cells.36  Overall, similarly to GEP studies, total concordance has not been achieved among the microRNA signatures from separate studies. Nevertheless, the up-regulation of microRNAs transcribed from genes located at 14q32 in APL with t(15;17) and the down-regulation of miR-133a in CBF-AML with t(8;21) were found in 2 or more studies (Table 1).21,36,37 

Table 1

Most significant microRNAs associated with cytogenetics and molecular features in AML

Chromosome aberrationmicroRNA
Up-regulatedDown-regulated
t(15;17)21,36,37  miR-127 miR-17 
 miR-299 miR-126 
 miR-323  
 miR-368  
 miR-382  
t(8;21)21,23,36,37  miR-126/miR-126miR-133 
inv(16)   
t(11q23)/MLL21,22,36-38  miR-326 miR-29s 
 miR-219 miR-34a 
 miR-17-92 miR-16 
 miR-196a  
Trisomy 822 miR-124a  
 miR-30d  
Cytogenetically normal   
    Gene alteration   
        NPM1 mutation21,39-41  miR-10a/-10b miR-204 
 miR-196a miR-128 
 miR-196b miR-126 
  miR-130a 
  miR-451 
        FLT3-ITD21-23,39  miR-155  
        CEBPA mutation21,42,43  miR-181a miR-34a 
 miR-335  
        BAALC overexpression44   miR-148a 
        MN1 overexpression45  miR-126 miR-16 
 miR-126miR-17-92 
 miR-424 miR-100 
  miR-196a 
Chromosome aberrationmicroRNA
Up-regulatedDown-regulated
t(15;17)21,36,37  miR-127 miR-17 
 miR-299 miR-126 
 miR-323  
 miR-368  
 miR-382  
t(8;21)21,23,36,37  miR-126/miR-126miR-133 
inv(16)   
t(11q23)/MLL21,22,36-38  miR-326 miR-29s 
 miR-219 miR-34a 
 miR-17-92 miR-16 
 miR-196a  
Trisomy 822 miR-124a  
 miR-30d  
Cytogenetically normal   
    Gene alteration   
        NPM1 mutation21,39-41  miR-10a/-10b miR-204 
 miR-196a miR-128 
 miR-196b miR-126 
  miR-130a 
  miR-451 
        FLT3-ITD21-23,39  miR-155  
        CEBPA mutation21,42,43  miR-181a miR-34a 
 miR-335  
        BAALC overexpression44   miR-148a 
        MN1 overexpression45  miR-126 miR-16 
 miR-126miR-17-92 
 miR-424 miR-100 
  miR-196a 

Distinct patterns of microRNA expression in AML with t(11q23)/MLL were reported.22  Garzon et al22  identified 8 microRNAs up-regulated (miR-326, miR-219, miR-194, miR-301, miR-324, miR-339, miR-99b, and miR-328) and 14 down-regulated (miR-34b, miR-15a, miR-29a, miR-29c, miR-372, miR-30a, miR-29b, miR-30e, miR-196a, let-7f, miR-102, miR-331, miR-299, and miR-193) in patients with t(11q23)/MLL versus all other AML patients. Many of the microRNAs down-regulated in t(11q23)/MLL-positive patients target critical oncogenes, for instance, miR-34b targets (→) CDK4 and CCNE2, miR-15aBCL2, the let-7 family → RAS, the miR-29 family → DNMT3A, DNMT3B, MCL1, and TCL1, miR-372LATS2.22  In another study,36  AML with t(11q23)/MLL was characterized by significant overexpression of miR-196b and 7 microRNAs from a unique polycistronic microRNA cluster miR-17-92: miR-17-5p, miR-17-3p, miR-18a, miR-19a, miR-19b, miR-20a, and miR-92. A follow-up report46  demonstrated that aberrant overexpression of the miR-17-92 cluster was caused by its up-regulation by the MLL gene fusions via their direct binding to the promoter region of C13orf25, the miR-17-92 host gene, and the resulting chromatin modification. In addition, the miR-17-92 cluster was up-regulated as a result of DNA amplification of the 13q31 locus, where the cluster is located. Mi et al46  identified 363 potential target genes of miR-17-92 whose expression was inversely correlated with the microRNAs expression and, using gene ontology, found that these genes were significantly enriched in cell differentiation (myeloid and B-cell differentiation), cell cycle, hematopoiesis, and cell death and apoptosis, suggesting that miR-17-92 contributes to leukemogenesis by down-regulating target genes promoting cell differentiation and apoptosis and those inhibiting cell proliferation. Recently, CDKN1A (p21) was identified as a direct target through which miR-17-92 polycistron modulates cell cycle and differentiation status, and self-renewal of t(11q23)/MLL leukemia stem cells.47 

The t(11q23)/MLL-rearranged AML category is very heterogeneous cytogenetically, with more than 50 different translocation partners of 11q23/MLL characterized at the molecular level to date.48  Preliminary data show that this heterogeneity also characterizes microRNA expression. When patients with t(6;11)(q27;q23) were compared with those with t(9;11)(p22;q23), 16 microRNAs were up-regulated in patients with t(6;11), including the antiapoptotic miR-21, which targets the tumor suppressor PTEN, and miR-26a and miR-26b, which target the TGFB1 regulator SMAD1.49,50  Studies on other specific t(11q23)/MLL rearrangements are needed to further characterize their microRNA patterns.

Garzon et al22  identified a signature composed of 42 up-regulated and no down-regulated microRNAs in patient samples with isolated trisomy 8 (+8), which were compared with AML patients with other karyotypes that did not contain secondary +8. Among the up-regulated microRNAs, miR-124a and miR-30d are located at 8p21 and 8q23, respectively, suggesting that a gene dosage effect may play a role in their up-regulation. Interestingly, miR-124a targets the myeloid transcription factor C/EBPα.51  In contrast, Dixon-McIver et al37  did not find correlation between +8 and microRNA expression, but they studied samples with an additional +8 rather than those with +8 as a sole cytogenetic abnormality.

Finally, a signature composed of 10 up-regulated (miR-10a, miR-10b, miR-26a, miR-30c, let-7a-2, miR-16-2, miR-21, miR-181b, miR-368, and miR-192) and 13 down-regulated microRNAs (miR-126, miR-203, miR-200c, miR-182, miR-204, miR-196b, miR-193, miR-191, miR-199a, miR-194, miR-183, miR-299, and miR-145) was identified in CN-AML.22  In the same subset of patients, miR-10a, miR-10b, and miR-196a-1 were correlated with expression of homeobox (HOX) genes. This is consistent with a high incidence of NPM1 mutations in CN-AML, between 46% and 62%,52  and the reported NPM1 mutation-associated gene-expression signature known to encompass HOX gene overexpression.53  However, as discussed in “Correlations of microRNA expression with molecular markers in CN-AML,” CN-AML consists of several molecular subsets characterized by the presence of recurrent gene mutations and expression changes. Hence, instead of a single microRNA-expression signature, CN-AML is rather associated with several microRNA-expression signatures denoting particular gene alterations occurring in this cytogenetic category.

Although microRNA-profiling appears to differentiate among distinct cytogenetic groups, the specific signatures differ among studies (Table 1). This is probably the result of the lack of standardization of the analytic methods used by different groups and currently precludes using microRNA-expression profiles as a diagnostic criterion. Nevertheless, microRNA profiling might become a diagnostic tool because the stability of microRNAs over time is better than that of longer, coding mRNAs used in GEP analyses, and the diagnostic accuracy of microRNA profiling might also be better. For example, using a combination of any 2 of a set of 4 microRNAs, it was possible to discriminate ALL from AML cases with an overall diagnostic accuracy of 97% to 98%.20  This was dramatically different from the results obtained with GEP, where a much larger number of probes were necessary to classify ALL and AML patients correctly.25 

Although most AML patients harbor at least one chromosomal aberration at diagnosis, 40% to 49% of AML patients have a normal karyotype and thus constitute the largest single cytogenetic subset of AML.29  During the past 15 years, CN-AML has been found to be very heterogeneous molecularly, with several gene mutations and gene expression alterations demonstrated to affect clinical outcome of CN-AML patients.52,54  Molecular alterations shown repeatedly to confer adverse prognosis include an internal tandem duplication of the FLT3 gene (FLT3-ITD),54-58  partial tandem duplication of the MLL gene (MLL-PTD),54,59  mutations of the WT1,60-62 IDH1 and IDH2 genes,63,64  and high expression of the BAALC,44 ERG,65  and MN145,66  genes, whereas favorable prognosis is bestowed by mutations in the NPM140,54,67,68  and CEBPA genes42,54 ; in the case of the latter, especially biallelic mutations.69,70  Moreover, combinations of markers have been found useful for outcome prediction of CN-AML patients. For example, patients with NPM1 mutations but no FLT3-ITD have a significantly better outcome than patients who harbor FLT3-ITD (with or without NPM1 mutations) or have FLT3 and NPM1 wild-type alleles.67 

Recently, several of the aforementioned recurrent molecular rearrangements in CN-AML have been correlated with microRNA- expression signatures (Table 1).

Mutations of the NPM1 gene have been linked to a distinctive microRNA-expression signature that includes the up-regulation of miR-10a, miR-10b, and miR-196a,39  all of which reside in the genomic cluster of HOX genes. Notably, HOX gene family up-regulation is a prominent characteristic of gene-expression signatures identified in patients with NPM1 mutations.53  In addition, Garzon et al39  reported down-regulation of miR-204 and miR-128a in AML with NPM1 mutations and showed in cell line experiments that miR-204 inhibited expression of MEIS1 and HOXA10, members of the HOX gene cluster. Recently, Becker et al40  reported a microRNA-expression signature, composed of 68 microRNAs, associated with NPM1 mutations in patients 60 years of age or older with de novo AML. Thirty-four microRNA-probes were up-regulated and 34 down-regulated in NPM1 mutated patients. Similar to previous studies, the prominent feature of the signature associated with NPM1 mutations was up-regulation of microRNAs embedded in the HOX cluster, miR-10a, miR-10b, miR-196a, and miR-196b.40  Down-regulated were miR-126, whose expression was recently positively correlated with MN1 expression in younger CN-AML patients,45  and miR-130a and miR-451, which are involved in, respectively, megakaryocytic differentiation and erythropoiesis.6  Importantly, NPM1 mutation status could be predicted with high accuracy based on microRNA-expression profiles. In leave-one-out cross-validated analysis, the mutation status of 92.5% of patients was correctly predicted (sensitivity = 96.2%; specificity = 87.5%).40 

Two groups reported independently that miR-155 was up-regulated in AML patients with FLT3-ITD, suggesting that miR-155 contributes to the highly proliferative phenotype of this molecular subset of AML.21,22  This hypothesis is supported by data from animal models showing that forced miR-155 expression can drive granulocyte/monocyte expansion, resulting in myeloid proliferation with dysplastic changes.71 MiR-155 was also the most overexpressed microRNA in the first FLT3-ITD–associated microRNA-expression signature derived in primary CN-AML patients 60 years of age or older that encompassed 32 differentially expressed probes.58  In addition, overexpressed in this signature was miR-125b-2*, and underexpressed were miR-144 and miR-451, normally up-regulated during erythropoiesis,72  and miR-488 and miR-486–5p; the latter is underexpressed in multiple cancers.73 

CEBPA mutations have been associated with an up-regulation of miR-181a and miR-335 in 2 AML studies,21,42  one of which was performed exclusively on samples from CN-AML patients.42  Notably, in this study,42 CEBPA mutations were associated with up-regulation of several genes involved in erythroid differentiation, in agreement with the results by Choong et al74  reporting an increase of miR-181a and miR-181b levels during erythroid differentiation. Consequently, high expression of the miR-181 family members seems to contribute to the partial erythroid differentiation of leukemic blasts harboring CEBPA mutations.42  Down-regulated in CEBPA-mutated patients were miR-194 and miR-34a.42 MiR-34a was recently shown to be a target of C/EBPα during granulopoiesis, and its low expression in leukemic blasts with CEBPA mutations was demonstrated to elevate levels of E2F3 and its transcriptional target, E2F1, which in turn increased proliferation and inhibited granulocytic differentiation of the blasts.43  Forced expression of miR-34a resulted in granulocytic differentiation of AML blasts carrying CEBPA mutations, suggesting that increasing miR-34a levels might become useful therapeutically,43  as could manipulation of expression of miR-223, which is part of a molecular network regulating granulopoiesis that also includes E2F1 and C/EBPα.75  Finally, Hackanson et al51  showed that CEBPA mRNA is targeted by miR-124a in vitro, and that miR-124a is epigenetically silenced in leukemia cell lines. Epigenetic treatment with the DNA-demethylating agent 5-aza-2′deoxycytidine caused up-regulation of miR-124a.51 

Our group has recently demonstrated the existence of a striking inverse correlation between BAALC expression and expression of miR-148a in younger CN-AML patients.44  This suggests that miR-148a might act as a negative regulator of BAALC expression and that low or absent miR-148a expression may increase expression of BAALC and the resulting aggressive phenotype.44  In patients 60 years of age or older, we derived a microRNA-expression signature composed of 18 microRNAs, 10 up-regulated and 8 down-regulated, in low BAALC-expressing patients.76  Consistent with higher expression of HOX genes in low BAALC-expressers, microRNAs embedded in the HOX cluster, miR-10a, miR-10b, and miR-9, were up-regulated in low BAALC expressers, whereas underexpressed were miR-126, positively correlated with MN1 expression (see the next paragraph), and miR-222, linked with hematologic lineage differentiation and known to target KIT.76 

We have also reported an MN1-associated microRNA-expression signature composed of 15 microRNAs (Table 1).45  Of those, expression of 8 microRNAs was positively, and expression of 7 negatively, correlated with MN1 expression. Five of the 8 microRNA probes positively associated with MN1 expression were members of the miR-126 family (including miR-126 and miR-126*). This microRNA family was recently reported to enhance the proangiogenic activity of vascular endothelial growth factor and regulate new blood vessel formation.77  We also noted up-regulation of miR-424, a regulator of monocyte and macrophage differentiation.78  Among the microRNAs negatively correlated with MN1, we found microRNAs involved in apoptosis (miR-16),79  malignant transformation (miR-19a and miR-20a, members of the miR-17-92 polycistron),11  or regulation of ERG expression (miR-196a).41 

Schwind et al76  reported recently the first microRNA-expression signature associated with ERG expression. The signature consisted of 11 microRNAs, 5 up-regulated and 6 down-regulated, in low ERG expressers. Among the former was miR-107, targeting NFIX, a gene involved in a regulatory feedback loop involving miR-223 and CEBPA during granulocytic differentiation,80 miR-148a, and miR-208, predicted in silico to target ERG itself. Down-regulated was miR-302d, a microRNA associated with early developmental stages and “stemness.”76 

To our knowledge, only one study has derived a microRNA-expression signature associated with R172 mutation in IDH2.63  Among the most up-regulated microRNAs in patients with R172 IDH2 mutations were miR-1 and miR-133, hitherto not associated with human cancer but involved in embryonal stem-cell differentiation,81  and miR-125a-5p and miR-125b, which target TP53. MiR-125b-1 is strongly up-regulated in AML with t(2;11)(p21;q23) and inhibits myeloid differentiation,82  whereas miR-125b-2 cooperates with GATA1 mutations during leukemogenesis of Down syndrome–associated acute megakaryoblastic leukemia.83  Moreover, overexpression of both miR-125b-1 and miR-125b-2 causes a dose-dependent myeloproliferative disorder progressing to a lethal myeloid leukemia in mice.84  None of the most down-regulated microRNAs in R172 IDH2-mutated patients, miR-194-1, miR-526, miR-520a-3p, or miR-548b, has yet been associated with AML, although related miR-520h promotes hematopoietic stem cell differentiation.8  Importantly, the microRNA-expression signature appeared to predict the R172 IDH2 mutational status with high accuracy, suggesting that patients with R172 IDH2 mutations differ biologically from patients with wild-type IDH2.63 

Debernardi et al85  were the first to demonstrate that miR-181a was more highly expressed in leukemic blasts with FAB M1 and M2 marrow morphology compared with FAB M4 and M5. A subsequent study confirmed these results with regard to both miR-181a and miR-181b.86  Another study,87  comparing the microRNA expression profiles of FAB M1 and FAB M5 subtypes, found the higher expression of not only miR-181a and miR-181b but also miR-181a*, miR-181d, miR130a, miR-135b, miR-146a, miR-146b, and miR-663 in FAB M1. In FAB M5 samples, miR-21, miR-193a, and miR-370 were overexpressed. Lutherborrow et al87  demonstrated significant down-regulation of miR-181a, miR-181b, miR-181d, miR130a, miR-135b, and miR-146a during induced monocytic differentiation of AML cell lines, suggesting direct involvement of these microRNAs in monocytic differentiation. They also identified key myeloid factors, such as MAFB, IRF8, and KLF4, as targets of these microRNAs.87 

Recent studies have shown that changes in microRNA expression can influence clinical outcome of AML patients. Dixon-McIver et al37  correlated microRNA expression with cytogenetic-risk groups: patients with favorable cytogenetic findings [ie, t(8;21), inv(16) and t(15;17)] had low expression of miR-let7b and miR-9, whereas high expression of these microRNAs was detected in samples from patients classified in the adverse or intermediate cytogenetic-risk groups.

Garzon et al22  reported that in a cytogenetically heterogeneous cohort of 122 newly diagnosed AML patients with predominantly intermediate- and poor-risk cytogenetics, overexpression of miR-20a, miR-25, miR-191, miR-199a, and miR-199b, assessed using microRNA microarrays, adversely affected overall survival. Validation of these results in a separate cohort of AML patients using a different profiling method (real-time reverse-transcription polymerase chain reaction) confirmed that high expression levels of miR-191 and miR-199a conferred inferior event-free and overall survival. In multivariable analyses, the 2 microRNAs remained independently predictive of both survival endpoints after adjustment for cytogenetics.22 

A recent Cancer and Leukemia Group B (CALGB) study identified a microRNA-expression signature with prognostic significance in CN-AML patients with high-risk molecular features: FLT3-ITD, wild-type NPM1 alleles, or both.88  The signature, composed of 12 microRNA probes derived from a training set of patients treated on CALGB protocol 19808, was associated with event-free survival. It was afterward tested in a validation set of similarly treated patients enrolled onto CALGB protocol 9621. Increased expression of 5 probes representing miR-181a and miR-181b was associated with decreased risk of an event (failure to achieve complete remission, relapse, or death), whereas higher expression of miR-124, miR-128-1, miR-194, miR-219-5p, miR-220a, and miR-320 associated with an increased risk.

In addition, the genome-wide microRNA-expression profile was integrated with a gene-expression signature to identify genes regulated by microRNAs whose altered expression contributed to leukemogenesis in CN-AML with molecular high-risk features.88  Expression of 452 genes significantly correlated with the prognostic microRNA-expression signature and included genes encoding proteins involved in innate immunity, such as intracellular sensors of microbial components and cell injury, which are part of the “inflammasome” regulating activation of caspase-1 and interleukin-1β.89  Among these genes, TLR4, CARD12, CASP1, and IL1B were predicted targets of members of the miR-181 family, and their expression was inversely correlated with the expression levels of miR-181 family members. These data support a functional relationship between microRNA and gene expression and suggest that down-regulation of members of the miR-181 family may contribute to the aggressive leukemia phenotype.88  Conversely, high expression of miR-181 family members may reduce the aggressiveness of the disease. The latter was confirmed by a subsequent study showing that up-regulated miR-181a and miR-181b were part of a microRNA-expression signature associated with prognostically favorable CEBPA mutations.42 

The aforementioned data suggest that measurements of microRNA expression may be useful for risk assessment in CN-AML. However, microRNA expression has been determined in these studies by genome-wide microarray profiling, which is relatively difficult to apply for prospective assessment of the molecular risk in individual patients. Therefore, Schwind et al89  investigated whether expression of a single microRNA, miR-181a, could provide independent prognostic information. Indeed, higher expression of miR-181a was found to be associated with a higher complete remission rate, longer overall survival, and a trend for longer disease-free survival in all CN-AML patients studied, and with higher complete remission rates and longer overall and disease-free survival in a subset with high-risk molecular features. Importantly, in both patient cohorts, higher miR-181a expression was significantly associated with better outcome in multivariable analyses. These data suggest that miR-181a expression may refine molecular risk classification of CN-AML and be used for molecular risk assessment in individual patients. Moreover, novel therapeutic options might be developed, such as delivery of miR-181a directly to the leukemia cells, or the use of agents capable of increasing endogenous levels of miR-181a, which might benefit patients with prognostically adverse low miR-181a expression.90 

Over the past 3 years, several studies have defined patterns of microRNA expression associated with cytogenetics, molecular subgroups, and clinical outcome in AML. Current efforts are now concentrated on performing biologic validation of these findings. Many questions remain unanswered. Do microRNAs contribute to myeloid leukemogenesis? If they do, what are the mechanisms? In the next 4 sections, we discuss the currently most relevant microRNAs involved in myeloid leukemogenesis (Table 2).

Table 2

MicroRNAs with a documented functional role in myeloid leukemogenesis

microRNALocationExpressionRegulated byTargetsIn vitro effectsIn vivo effects
miR-155 21q21.3 Up-regulated in FLT3-ITD21,22,39  NFκB8  SHIP1, CEBPB91,92  Blocks megakaryopoiesis93  Induces myeloid colonies Induces myeloproliferation with MDS changes in mice71  
miR-196b 7p15 Up-regulated in t(11q23)/MLL36,38  MLL38  HOXB8 Increases cell survival and proliferation of progenitors38   
miR-223 Xq12 Down-regulated in t(8;21)94  RUNX1/RUNX1T1, CEBPA, NFIA1,94  MEF2C95  Induces granulocytic differentiation95  Granulocytosis in miR-223 KO mice95  
miR-29b-1 7q32 Down-regulated in wild-type NPM1, t(11q23)/MLL and −7 and del(7q)22,96,97  NFκB, MYC, YY16  MCL1, CDK6, SP1, DNMT3A/B13,97,98  Induces apoptosis97,98  Inhibits proliferation97,98  Regulates negatively DNA methylation13  Reduces tumorigenicity97  
microRNALocationExpressionRegulated byTargetsIn vitro effectsIn vivo effects
miR-155 21q21.3 Up-regulated in FLT3-ITD21,22,39  NFκB8  SHIP1, CEBPB91,92  Blocks megakaryopoiesis93  Induces myeloid colonies Induces myeloproliferation with MDS changes in mice71  
miR-196b 7p15 Up-regulated in t(11q23)/MLL36,38  MLL38  HOXB8 Increases cell survival and proliferation of progenitors38   
miR-223 Xq12 Down-regulated in t(8;21)94  RUNX1/RUNX1T1, CEBPA, NFIA1,94  MEF2C95  Induces granulocytic differentiation95  Granulocytosis in miR-223 KO mice95  
miR-29b-1 7q32 Down-regulated in wild-type NPM1, t(11q23)/MLL and −7 and del(7q)22,96,97  NFκB, MYC, YY16  MCL1, CDK6, SP1, DNMT3A/B13,97,98  Induces apoptosis97,98  Inhibits proliferation97,98  Regulates negatively DNA methylation13  Reduces tumorigenicity97  

Human miR-155, mapped to 21q21.3, resides in the noncoding BIC transcript, and its expression is elevated in many solid tumors, lymphomas, and acute leukemias.6,9,21,22,99  In AML, miR-155 expression is higher in patients carrying FLT3-ITD mutations.21,22,39  However, blocking FLT3 phosphorylation activity with a FLT3 inhibitor or overexpressing FLT3-ITD in mouse myeloid precursor cells did not change miR-155 expression, suggesting that miR-155 expression is independent from FLT3-ITD signaling.39  Functionally, overexpression of miR-155 in human CD34+ stem cells results in myeloid proliferation with blockage of erythroid and megakaryocytic colony formation.93  Moreover, enforced up-regulation of miR-155 in normal murine hematopoietic stem cells induced myeloproliferation, although overt AML was not observed, indicating that transformation to an overt AML requires cooperation of additional genetic alterations.91  In contrast, the ectopic up-regulation of miR-155 in mouse lymphocyte precursors induced not only polyclonal lymphocytosis but was also sufficient to induce high-grade leukemia/lymphoma.92  However, the mechanistic basis for miR-155 biologic effects on the myeloid and lymphoid lineages remains largely unresolved. Recently, 2 independent reports identified Src homology-2 domain-containing inositol 5-phosphatase 1 (SHIP1) as a direct target of miR-155.91,92  SHIP1 is a negative regulator of cell signaling in the immune system.100  This phosphatase is also implicated in B-cell maturation because it has a differential expression in the pro-B compared with the pre-B stage.100  Mice with global SHIP1 deficiency develop a myeloproliferative disease characterized by increased granulocyte-monocyte populations and decreased B-lymphocyte numbers.101  Thus, SHIP1-deficient mice resemble the phenotype of miR-155 transgenic mice. Interestingly, SHIP1 is mutated in a small subset of AML patients, in whom loss of function of SHIP1 has been implicated in the oncogenic process.102  In addition to SHIP1, miR-155 targets CEBPB,92  encoding a protein with a critical function in granulopoiesis,103  suggesting that leukemogenic function of miR-155 results in deregulation of SHIP1 and CEBPB (Table 2).91,92 

miR-196b, located between HOXA9 and HOXA10 genes at 7p15, has been found up-regulated in AML patients with t(11q23)/MLL.21,22,36-38  Popovic et al38  demonstrated that miR-196b overexpression in BM progenitor cells resulted in a partial block of their differentiation and increased proliferative capacity and survival, and that expression of miR-196b was induced by leukemogenic MLL fusion proteins. Importantly, treatment of BM cells transformed with the MLL-AF9 fusion gene with the antagomir targeting miR-196b decreased proliferative capacity of the cells (Table 2).38  This suggests that miR-196b might become a novel therapeutic target in AML with t(11q23)/MLL.

miR-223, located at Xq12, is expressed at low levels in hematopoietic stem cells, but its expression increases dramatically during granulocytic differentiation.6  Hence, it was suggested that miR-223 induces granulocytic differentiation. To answer this question, Johnnidis et al95  engineered a miR-223 knockout mouse and found, rather surprisingly, that these mice exhibit granulocytosis and a hyperinflamatory state. Further experiments demonstrated that miR-223 exhibits its antiproliferative effects in granulopoiesis by targeting the transcription factor MEF2C. A recent study reported that MEF2C mRNA levels are up-regulated in highly proliferative leukemic granulocyte-monocyte progenitors and that MEF2C ectopic expression enhanced their proliferation.104  Thus, miR-223 acts normally as a negative regulator of granulocytic proliferation (Table 2) as part of an autoregulatory negative feedback loop also involving C/EBPα and E2F1.75  It has been reported that AML patients with t(8;21) exhibit lower miR-223 levels than other AMLs.94  Furthermore, miR-223 was shown to be a direct transcriptional target of RUNX1/RUNX1T1 through the recruitment of chromatin remodeling enzymes. Treatment of AML blasts with hypomethylating agents, RUNX1/RUNX1T1 inhibitors, or miR-223 oligonucleotides enhanced miR-223 levels and restored cell differentiation.94 

The miR-29 family is composed of 3 isoforms arranged in 2 clusters: miR-29b-1/miR-29a located at 7q32 and miR-29b-2/miR-29c located at 1q23. Interestingly, band 7q32 is frequently deleted in AML and myelodysplastic syndromes with del(7q).26 MiR-29 family members are down-regulated in high-risk chronic lymphocytic leukemia, lung cancer, invasive breast cancer, and cholangiocarcinoma.98,105-107  We and others reported down-regulation of miR-29 family members in primary AML samples with t(11q23)/MLL22,97  and those with del(7q),22,96,97  whereas miR-29a expression was increased in AML samples from (mostly) FLT3-ITD–negative CN-AML patients.108  Moreover, lower levels of miR-29 were detected in CN-AML samples with wild-type NPM1 compared with CN-AML with NPM1 mutations.39  Recently, ectopic overexpression of miR-29b in AML cell lines and primary AML blasts was shown to induce apoptosis and inhibit blast proliferation.97  Furthermore, direct inoculation of synthetic miR-29b oligonucleotides into xenograft tumors dramatically decreased tumor growth (Table 2). By analyzing mRNA expression changes in AML cell lines after miR-29a and miR-29b overexpression, several targets were identified, including oncogenes MCL1, CDK6, IGFR, and JAK2. Notably, antiapoptotic genes, including MCL1, were enriched among the mRNAs inversely correlated with miR-29 expression (Table 2).97  Our group also reported that miR-29b modulates DNA methylation by targeting directly DNMT3a and DNMT3b and indirectly DNMT1 through SP1.13  Blum et al109  observed that those AML patients 60 years of age or older treated with single-agent decitabine, a DNMT inhibitor, who responded to treatment had higher pretreatment levels of miR-29b. Moreover, a recent study15  showed that miR-29b is involved in a protein-microRNA network, which includes SP1, NFκB, and HDAC, whose deregulation results in KIT overexpression in AML and is associated with adverse clinical outcome. Treatment with ectopic miR-29b or compounds capable of increasing levels of endogenous miR-29b (eg, bortezomib, mithramycin A, or bay11-7082) resulted in KIT down-regulation.15  These data support a tumor suppressor function of miR-29b and provide a rationale for the use of synthetic miR-29b oligonucleotides or agents increasing miR-29b expression as novel therapeutic options in AML.

The development and increasing use of high-throughput genome-scale technologies are changing the way we approach the study of leukemia. By performing array-based comparative genomic hybridization, single nucleotide polymorphism array analyses, mRNA- and microRNA-expression profiling studies, and whole genome sequencing, it is possible to attain a comprehensive, multidimensional view of leukemia. Knowledge gained from these approaches is critical to better understanding of leukemogenesis and development of novel therapies. Two principal challenges to this approach exist. The first concerns technical problems inherent in the platforms used to analyze biologic samples, such as reproducibility, sensitivity, and background noise. The second relates to difficulty of processing large amounts of data and includes standardization in the analysis and reporting of results. Finally, this comprehensive integrative analysis will require experimental validation. In particular, in vivo studies using transgenic and conditional “knockout” or “knockin” animal models should be performed.

The emergence of novel technologies, especially high-throughput small RNA sequencing methods, will allow more sensitive and efficient microRNA detection in patient samples,110,111  and identification of novel microRNAs.112  In addition, sequence information will be available, and mutations or polymorphisms could be discovered. However, standardization of these new methods is necessary to overcome the variability observed when different microRNA-expression detection platforms are used.

There is also the potential to develop microRNA-based therapeutic strategies.113,114  It is reasonable to restore lost tumor suppressor microRNA in AML blasts, such as miR-29 or miR-181a, using synthetic oligonucleotides that mimic the endogenous mature microRNA or compounds that indirectly up-regulate microRNA expression. For example, miR-223 and miR-29b expression could be restored in RUNX1/RUNX1T1-positive blasts using hypomethylating agents, such as decitabine. Conversely, silencing a highly expressed microRNA, such as miR-155 or miR-196a, with antisense oligonucleotides against mature microRNA sequences could also provide clinical benefit. Preliminary data indicate that using antagomirs is an effective approach to silence microRNA expression in mice.115  Elmén et al116  recently reported that the simple systemic delivery of an unconjugated locked-nucleic acid modified oligonucleotide (LNA-antimiR) effectively antagonized miR-122 expressed in the liver of nonhuman primates. Furthermore, LNA-antimiR-122 (SPC3649) is the first microRNA-targeted drug to enter human clinical trials; a phase 1 clinical trial testing the safety of intravenous administration of LNA-antimiR-122 in healthy volunteers is ongoing in Denmark (www.santaris.com).

This work was supported in part by the National Cancer Institute, Bethesda, MD (grants CA101140, CA16058, and CA102031), the Kimmel Cancer Research Foundation, the Coleman Leukemia Research Foundation, and the Harry T. Mangurian Jr Foundation.

National Institutes of Health

Contribution: G.M., K.M., M.D.R., R.G., and C.D.B. participated in writing this article and approved its final version.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Guido Marcucci, The Ohio State University, Comprehensive Cancer Center, Biomedical Research Tower, 460 West 12th Ave, Columbus, OH 43210; e-mail: guido.marcucci@osumc.edu; and Krzysztof Mrózek, The Ohio State University, Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Rm 1232A, 300 West 10th Ave, Columbus, OH 43210-1228; e-mail: krzysztof.mrozek@osumc.edu.

1
Ambros
 
V
The functions of animal microRNAs.
Nature
2004
, vol. 
431
 
7006
(pg. 
350
-
355
)
2
Bartel
 
DP
MicroRNAs: target recognition and regulatory functions.
Cell
2009
, vol. 
136
 
2
(pg. 
215
-
233
)
3
Lal
 
A
Navarro
 
F
Maher
 
CA
, et al. 
miR-24 inhibits cell proliferation by targeting E2F2, MYC, and other cell-cycle genes via binding to “seedless” 3′UTR microRNA recognition elements.
Mol Cell
2009
, vol. 
35
 
5
(pg. 
610
-
625
)
4
Vasudevan
 
S
Tong
 
Y
Steitz
 
JA
Switching from repression to activation: microRNAs can up-regulate translation.
Science
2007
, vol. 
318
 
5858
(pg. 
1931
-
1934
)
5
Griffiths-Jones
 
S
Grocock
 
RJ
van Dongen
 
S
Bateman
 
A
Enright
 
AJ
miRBase: microRNA sequences, targets and gene nomenclature.
Nucleic Acids Res
2006
, vol. 
34
 (pg. 
D140
-
D144
Database issue
6
Garzon
 
R
Croce
 
CM
MicroRNAs in normal and malignant hematopoiesis.
Curr Opin Hematol
2008
, vol. 
15
 
14
(pg. 
352
-
358
)
7
Vasilatou
 
D
Papageorgiou
 
S
Pappa
 
V
Papageorgiou
 
E
Dervenoulas
 
J
The role of microRNAs in normal and malignant hematopoiesis.
Eur J Haematol
2010
, vol. 
84
 
1
(pg. 
1
-
16
)
8
Havelange
 
V
Garzon
 
R
MicroRNAs: emerging key regulators of hematopoiesis.
Am J Hematol
2010
, vol. 
85
 
12
(pg. 
935
-
942
)
9
Calin
 
GA
Croce
 
CM
MicroRNA signatures in human cancers.
Nat Rev Cancer
2006
, vol. 
6
 
11
(pg. 
857
-
866
)
10
Calin
 
GA
Sevignani
 
C
Dumitru
 
CD
, et al. 
Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers.
Proc Natl Acad Sci U S A
2004
, vol. 
101
 
9
(pg. 
2999
-
3004
)
11
Xiao
 
C
Srinivasan
 
L
Calado
 
DP
, et al. 
Lymphoproliferative disease and autoimmunity in mice with increased miR-17-92 expression in lymphocytes.
Nat Immunol
2008
, vol. 
9
 
4
(pg. 
405
-
414
)
12
Costinean
 
S
Zanesi
 
N
Pekarsky
 
Y
, et al. 
Pre-B cell proliferation and lymphoblastic leukemia/high-grade lymphoma in Emu-miR-155 transgenic mice.
Proc Natl Acad Sci U S A
2006
, vol. 
103
 
18
(pg. 
7024
-
7029
)
13
Garzon
 
R
Liu
 
S
Fabbri
 
M
, et al. 
MicroRNA-29b induces global DNA hypomethylation and tumor suppressor gene reexpression in acute myeloid leukemia by targeting directly DNMT3A and 3B and indirectly DNMT1.
Blood
2009
, vol. 
113
 
25
(pg. 
6411
-
6418
)
14
Eiring
 
AM
Harb
 
JG
Neviani
 
P
, et al. 
miR-328 functions as an RNA decoy to modulate hnRNP E2 regulation of mRNA translation in leukemic blasts.
Cell
2010
, vol. 
140
 
5
(pg. 
652
-
665
)
15
Liu
 
S
Wu
 
L-C
Pang
 
J
, et al. 
Sp1/NFkappaB/HDAC/miR-29b regulatory network in KIT-driven myeloid leukemia.
Cancer Cell
2010
, vol. 
17
 
4
(pg. 
333
-
347
)
16
Garzon
 
R
Calin
 
GA
Croce
 
CM
MicroRNAs in cancer.
Annu Rev Med
2009
, vol. 
60
 (pg. 
167
-
179
)
17
Chen
 
J
Odenike
 
O
Rowley
 
JD
Leukaemogenesis: more than mutant genes.
Nat Rev Cancer
2010
, vol. 
10
 
1
(pg. 
23
-
36
)
18
Seca
 
H
Almeida
 
GM
Guimarães
 
JE
Vasconcelos
 
MH
miR signatures and the role of miRs in acute myeloid leukaemia.
Eur J Cancer
2010
, vol. 
46
 
9
(pg. 
1520
-
1527
)
19
Marcucci
 
G
Mrózek
 
K
Radmacher
 
MD
Bloomfield
 
CD
Croce
 
CM
MicroRNA expression profiling in acute myeloid and chronic lymphocytic leukaemias.
Best Pract Res Clin Haematol
2009
, vol. 
22
 
2
(pg. 
239
-
248
)
20
Mi
 
S
Lu
 
J
Sun
 
M
, et al. 
MicroRNA expression signatures accurately discriminate acute lymphoblastic leukemia from acute myeloid leukemia.
Proc Natl Acad Sci U S A
2007
, vol. 
104
 
50
(pg. 
19971
-
19976
)
21
Jongen-Lavrencic
 
M
Sun
 
SM
Dijkstra
 
MK
Valk
 
PJM
Löwenberg
 
B
MicroRNA expression profiling in relation to the genetic heterogeneity of acute myeloid leukemia.
Blood
2008
, vol. 
111
 
10
(pg. 
5078
-
5085
)
22
Garzon
 
R
Volinia
 
S
Liu
 
C-G
, et al. 
MicroRNA signatures associated with cytogenetics and prognosis in acute myeloid leukemia.
Blood
2008
, vol. 
111
 
6
(pg. 
3183
-
3189
)
23
Cammarata
 
G
Augugliaro
 
L
Salemi
 
D
, et al. 
Differential expression of specific microRNA and their targets in acute myeloid leukemia.
Am J Hematol
2010
, vol. 
85
 
5
(pg. 
331
-
339
)
24
Steidl
 
U
Kronenwett
 
R
Rohr
 
U-P
, et al. 
Gene expression profiling identifies significant differences between the molecular phenotypes of bone marrow-derived and circulating human CD34+ hematopoietic stem cells.
Blood
2002
, vol. 
99
 
6
(pg. 
2037
-
2044
)
25
Golub
 
TR
Slonim
 
DK
Tamayo
 
P
, et al. 
Molecular classification of cancer: class discovery and class prediction by gene expression monitoring.
Science
1999
, vol. 
286
 
5439
(pg. 
531
-
537
)
26
Mrózek
 
K
Heinonen
 
K
Bloomfield
 
CD
Clinical importance of cytogenetics in acute myeloid leukaemia.
Best Pract Res Clin Haematol
2001
, vol. 
14
 
1
(pg. 
19
-
47
)
27
Grimwade
 
D
Hills
 
RK
Moorman
 
AV
, et al. 
Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities amongst 5876 younger adult patients treated in the UK Medical Research Council trials.
Blood
2010
, vol. 
116
 
3
(pg. 
354
-
365
)
28
Mitelman
 
F
Johansson
 
B
Mertens
 
F
Mitelman database of chromosome aberrations and gene fusions in cancer.
Accessed August 27, 2010 
29
Mrózek
 
K
Heerema
 
NA
Bloomfield
 
CD
Cytogenetics in acute leukemia.
Blood Rev
2004
, vol. 
18
 
2
(pg. 
115
-
136
)
30
Ferrara
 
F
Palmieri
 
S
Leoni
 
F
Clinically useful prognostic factors in acute myeloid leukemia.
Crit Rev Oncol Hematol
2008
, vol. 
66
 
3
(pg. 
181
-
193
)
31
Döhner
 
H
Estey
 
EH
Amadori
 
S
, et al. 
Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet.
Blood
2010
, vol. 
115
 
3
(pg. 
453
-
474
)
32
Byrd
 
JC
Mrózek
 
K
Dodge
 
RK
, et al. 
Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461).
Blood
2002
, vol. 
100
 
13
(pg. 
4325
-
4336
)
33
Slovak
 
ML
Kopecky
 
KJ
Cassileth
 
PA
, et al. 
Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group study.
Blood
2000
, vol. 
96
 
13
(pg. 
4075
-
4083
)
34
Mrózek
 
K
Cytogenetic, molecular genetic, and clinical characteristics of acute myeloid leukemia with a complex karyotype.
Semin Oncol
2008
, vol. 
35
 
4
(pg. 
365
-
377
)
35
Farag
 
SS
Ruppert
 
AS
Mrózek
 
K
, et al. 
Outcome of induction and postremission therapy in younger adults with acute myeloid leukemia with normal karyotype: a Cancer and Leukemia Group B study.
J Clin Oncol
2005
, vol. 
23
 
3
(pg. 
482
-
493
)
36
Li
 
Z
Lu
 
J
Sun
 
M
, et al. 
Distinct microRNA expression profiles in acute myeloid leukemia with common translocations.
Proc Natl Acad Sci U S A
2008
, vol. 
105
 
40
(pg. 
15535
-
15540
)
37
Dixon-McIver
 
A
East
 
P
Mein
 
CA
, et al. 
Distinctive patterns of microRNA expression associated with karyotype in acute myeloid leukaemia.
PLoS ONE
2008
, vol. 
3
 
5
pg. 
e2141
 
38
Popovic
 
R
Riesbeck
 
LE
Velu
 
CS
, et al. 
Regulation of mir-196b by MLL and its overexpression by MLL fusions contributes to immortalization.
Blood
2009
, vol. 
113
 
14
(pg. 
3314
-
3322
)
39
Garzon
 
R
Garofalo
 
M
Martelli
 
MP
, et al. 
Distinctive microRNA signature of acute myeloid leukemia bearing cytoplasmic mutated nucleophosmin.
Proc Natl Acad Sci U S A
2008
, vol. 
105
 
10
(pg. 
3945
-
3950
)
40
Becker
 
H
Marcucci
 
G
Maharry
 
K
, et al. 
Favorable prognostic impact of NPM1 mutations in older patients with cytogenetically normal de novo acute myeloid leukemia and associated gene- and microRNA-expression signatures: a Cancer and Leukemia Group B study.
J Clin Oncol
2010
, vol. 
28
 
4
(pg. 
596
-
604
)
41
Coskun
 
E
von der Heide
 
EK
Schlee
 
C
, et al. 
The role of microRNA-196a and microRNA-196b as ERG regulators in acute myeloid leukemia and acute T-lymphoblastic leukemia [published online ahead of print June 4, 2010].
Leuk Res
 
42
Marcucci
 
G
Maharry
 
K
Radmacher
 
MD
, et al. 
Prognostic significance of, and gene and microRNA expression signatures associated with, CEBPA mutations in cytogenetically normal acute myeloid leukemia with high-risk molecular features: a Cancer and Leukemia Group B study.
J Clin Oncol
2008
, vol. 
26
 
31
(pg. 
5078
-
5087
)
43
Pulikkan
 
JA
Peramangalam
 
PS
Dengler
 
V
, et al. 
C/EBPalpha-regulated microRNA-34a targets E2F3 during granulopoiesis and is downregulated in AML with CEBPA mutations.
Blood
2010
, vol. 
116
 
25
(pg. 
5638
-
5649
)
44
Langer
 
C
Radmacher
 
MD
Ruppert
 
AS
, et al. 
High BAALC expression associates with other molecular prognostic markers, poor outcome, and a distinct gene-expression signature in cytogenetically normal patients younger than 60 years with acute myeloid leukemia: a Cancer and Leukemia Group B (CALGB) study.
Blood
2008
, vol. 
111
 
11
(pg. 
5371
-
5379
)
45
Langer
 
C
Marcucci
 
G
Holland
 
KB
, et al. 
Prognostic importance of MN1 transcript levels, and biologic insights from MN1-associated gene and microRNA expression signatures in cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study.
J Clin Oncol
2009
, vol. 
27
 
19
(pg. 
3198
-
3204
)
46
Mi
 
S
Li
 
Z
Chen
 
P
, et al. 
Aberrant overexpression and function of the miR-17-92 cluster in MLL-rearranged acute leukemia.
Proc Natl Acad Sci U S A
2010
, vol. 
107
 
8
(pg. 
3710
-
3715
)
47
Wong
 
P
Iwasaki
 
M
Somervaille
 
TC
, et al. 
The miR-17-92 microRNA polycistron regulates MLL leukemia stem cell potential by modulating p21 expression.
Cancer Res
2010
, vol. 
70
 
9
(pg. 
3833
-
3842
)
48
Meyer
 
C
Kowarz
 
E
Hofmann
 
J
, et al. 
New insights to the MLL recombinome of acute leukemias.
Leukemia
2009
, vol. 
23
 
8
(pg. 
1490
-
1499
)
49
Meng
 
F
Henson
 
R
Lang
 
M
, et al. 
Involvement of human micro-RNA in growth and response to chemotherapy in human cholangiocarcinoma cell lines.
Gastroenterology
2006
, vol. 
130
 
7
(pg. 
2113
-
2129
)
50
Lewis
 
BP
Shih
 
I
Jones-Rhoades
 
MW
Bartel
 
DP
Burge
 
CB
Prediction of mammalian microRNA targets.
Cell
2003
, vol. 
115
 
7
(pg. 
787
-
798
)
51
Hackanson
 
B
Bennett
 
KL
Brena
 
RM
, et al. 
Epigenetic modification of CCAAT/enhancer binding protein α expression in acute myeloid leukemia.
Cancer Res
2008
, vol. 
68
 
9
(pg. 
3142
-
3151
)
52
Mrózek
 
K
Marcucci
 
G
Paschka
 
P
, et al. 
Clinical relevance of mutations and gene-expression changes in adult acute myeloid leukemia with normal cytogenetics: are we ready for a prognostically prioritized molecular classification?
Blood
2007
, vol. 
109
 
2
(pg. 
431
-
448
)
53
Alcalay
 
M
Tiacci
 
E
Bergomas
 
R
, et al. 
Acute myeloid leukemia bearing cytoplasmic nucleophosmin (NPMc+ AML) shows a distinct gene expression profile characterized by up-regulation of genes involved in stem-cell maintenance.
Blood
2005
, vol. 
106
 
3
(pg. 
899
-
902
)
54
Schlenk
 
RF
Döhner
 
K
Krauter
 
J
, et al. 
Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia.
N Engl J Med
2008
, vol. 
358
 
18
(pg. 
1909
-
1918
)
55
Whitman
 
SP
Archer
 
KJ
Feng
 
L
, et al. 
Absence of the wild-type allele predicts poor prognosis in adult de novo acute myeloid leukemia with normal cytogenetics and the internal tandem duplication of FLT3: a Cancer and Leukemia Group B study.
Cancer Res
2001
, vol. 
61
 
19
(pg. 
7233
-
7239
)
56
Kottaridis
 
PD
Gale
 
RE
Frew
 
ME
, et al. 
The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials.
Blood
2001
, vol. 
98
 
6
(pg. 
1752
-
1759
)
57
Thiede
 
C
Steudel
 
C
Mohr
 
B
, et al. 
Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis.
Blood
2002
, vol. 
99
 
12
(pg. 
4326
-
4335
)
58
Whitman
 
SP
Maharry
 
K
Radmacher
 
MD
, et al. 
FLT3 internal tandem duplication associates with adverse outcome and gene- and microRNA-expression signatures in patients 60 years of age or older with primary cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study.
Blood
2010
, vol. 
116
 
18
(pg. 
3622
-
3626
)
59
Caligiuri
 
MA
Strout
 
MP
Lawrence
 
D
, et al. 
Rearrangement of ALL1 (MLL) in acute myeloid leukemia with normal cytogenetics.
Cancer Res
1998
, vol. 
58
 
1
(pg. 
55
-
59
)
60
Paschka
 
P
Marcucci
 
G
Ruppert
 
AS
, et al. 
Wilms' tumor 1 gene mutations independently predict poor outcome in adults with cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study.
J Clin Oncol
2008
, vol. 
26
 
28
(pg. 
4595
-
4602
)
61
Virappane
 
P
Gale
 
R
Hills
 
R
, et al. 
Mutation of the Wilms' tumor 1 gene is a poor prognostic factor associated with chemotherapy resistance in normal karyotype acute myeloid leukemia: the United Kingdom Medical Research Council Adult Leukaemia Working Party.
J Clin Oncol
2008
, vol. 
26
 
33
(pg. 
5429
-
5435
)
62
Becker
 
H
Marcucci
 
G
Maharry
 
K
, et al. 
Mutations of the Wilms tumor 1 gene (WT1) in older patients with primary cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study.
Blood
2010
, vol. 
116
 
5
(pg. 
788
-
792
)
63
Marcucci
 
G
Maharry
 
K
Wu
 
Y-Z
, et al. 
IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study.
J Clin Oncol
, vol. 
28
 
14
(pg. 
2348
-
2355
)
64
Paschka
 
P
Schlenk
 
RF
Gaidzik
 
VI
, et al. 
IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication.
J Clin Oncol
2010
, vol. 
28
 
22
(pg. 
3636
-
3643
)
65
Marcucci
 
G
Maharry
 
K
Whitman
 
SP
, et al. 
High expression levels of the ETS-related gene, ERG, predict adverse outcome and improve molecular risk-based classification of cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study.
J Clin Oncol
2007
, vol. 
25
 
22
(pg. 
3337
-
3343
)
66
Heuser
 
M
Beutel
 
G
Krauter
 
J
, et al. 
High meningioma 1 (MN1) expression as a predictor for poor outcome in acute myeloid leukemia with normal cytogenetics.
Blood
2006
, vol. 
108
 
12
(pg. 
3898
-
3905
)
67
Döhner
 
K
Schlenk
 
RF
Habdank
 
M
, et al. 
Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: interaction with other gene mutations.
Blood
2005
, vol. 
106
 
12
(pg. 
3740
-
3746
)
68
Schnittger
 
S
Schoch
 
C
Kern
 
W
, et al. 
Nucleophosmin gene mutations are predictors of favorable prognosis in acute myelogenous leukemia with a normal karyotype.
Blood
2005
, vol. 
106
 
12
(pg. 
3733
-
3739
)
69
Wouters
 
BJ
Löwenberg
 
B
Erpelinck- Eerschueren
 
CAJ
van Putten
 
WLJ
Valk
 
PJM
Delwel
 
R
Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that is uniquely associated with a favorable outcome.
Blood
2009
, vol. 
113
 
13
(pg. 
3088
-
3091
)
70
Dufour
 
A
Schneider
 
F
Metzeler
 
KH
, et al. 
Acute myeloid leukemia with biallelic CEBPA gene mutations and normal karyotype represents a distinct genetic entity associated with a favorable clinical outcome.
J Clin Oncol
2010
, vol. 
28
 
4
(pg. 
570
-
577
)
71
O'Connell
 
RM
Rao
 
DS
Chaudhuri
 
AA
, et al. 
Sustained expression of microRNA-155 in hematopoietic stem cells causes a myeloproliferative disorder.
J Exp Med
2008
, vol. 
205
 
3
(pg. 
585
-
594
)
72
Papapetrou
 
EP
Korkola
 
JE
Sadelain
 
M
A genetic strategy for single and combinatorial analysis of miRNA function in mammalian hematopoietic stem cells.
Stem Cells
2010
, vol. 
28
 
2
(pg. 
287
-
296
)
73
Navon
 
R
Wang
 
H
Steinfeld
 
I
Tsalenko
 
A
Ben-Dor
 
A
Yakhini
 
Z
Novel rank-based statistical methods reveal microRNAs with differential expression in multiple cancer types.
PLoS ONE
2009
, vol. 
4
 
11
pg. 
e8003
 
74
Choong
 
ML
Yang
 
HH
McNiece
 
I
MicroRNA expression profiling during human cord blood-derived CD34 cell erythropoiesis.
Exp Hematol
2007
, vol. 
35
 
4
(pg. 
551
-
564
)
75
Pulikkan
 
JA
Dengler
 
V
Peramangalam
 
PS
, et al. 
Cell-cycle regulator E2F1 and microRNA-223 comprise an autoregulatory negative feedback loop in acute myeloid leukemia.
Blood
2010
, vol. 
115
 
9
(pg. 
1768
-
1778
)
76
Schwind
 
S
Marcucci
 
G
Maharry
 
K
, et al. 
BAALC and ERG expression levels are associated with outcome and distinct gene- and microRNA-expression profiles in older patients with de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study.
Blood
2010
, vol. 
116
 
25
(pg. 
5660
-
5669
)
77
Wang
 
S
Aurora
 
AB
Johnson
 
BA
, et al. 
The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis.
Dev Cell
2008
, vol. 
15
 
2
(pg. 
261
-
271
)
78
Rosa
 
A
Ballarino
 
M
Sorrentino
 
A
, et al. 
The interplay between the master transcription factor PU.1 and miR-424 regulates human monocyte/macrophage differentiation.
Proc Natl Acad Sci U S A
2007
, vol. 
104
 
50
(pg. 
19849
-
19854
)
79
Cimmino
 
A
Calin
 
GA
Fabbri
 
M
, et al. 
miR-15 and miR-16 induce apoptosis by targeting BCL2.
Proc Natl Acad Sci U S A
2005
, vol. 
102
 
39
(pg. 
13944
-
13949
)
80
Fazi
 
F
Rosa
 
A
Fatica
 
A
, et al. 
A minicircuitry comprised of microRNA-223 and transcription factors NFI-A and C/EBPalpha regulates human granulopoiesis.
Cell
2005
, vol. 
123
 
5
(pg. 
819
-
831
)
81
Takaya
 
T
Ono
 
K
Kawamura
 
T
, et al. 
MicroRNA-1 and microRNA-133 in spontaneous myocardial differentiation of mouse embryonic stem cells.
Circ J
2009
, vol. 
73
 
8
(pg. 
1492
-
1497
)
82
Bousquet
 
M
Quelen
 
C
Rosati
 
R
, et al. 
Myeloid cell differentiation arrest by miR-125b-1 in myelodysplastic syndrome and acute myeloid leukemia with the t(2;11)(p21;q23) translocation.
J Exp Med
2008
, vol. 
205
 
11
(pg. 
2499
-
2506
)
83
Klusmann
 
J-H
Li
 
Z
Böhmer
 
K
, et al. 
miR-125b-2 is a potential oncomiR on human chromosome 21 in megakaryoblastic leukemia.
Genes Dev
2010
, vol. 
24
 
5
(pg. 
478
-
490
)
84
O'Connell
 
RM
Chaudhuri
 
AA
Rao
 
DS
Gibson
 
WSJ
Balazs
 
AB
Baltimore
 
D
MicroRNAs enriched in hematopoietic stem cells differentially regulate long-term hematopoietic output.
Proc Natl Acad Sci U S A
2010
, vol. 
107
 
32
(pg. 
14235
-
14240
)
85
Debernardi
 
S
Skoulakis
 
S
Molloy
 
G
Chaplin
 
T
Dixon-McIver
 
A
Young
 
BD
MicroRNA miR-181a correlates with morphological sub-class of acute myeloid leukaemia and the expression of its target genes in global genome-wide analysis.
Leukemia
2007
, vol. 
21
 
5
(pg. 
912
-
916
)
86
Isken
 
F
Steffen
 
B
Merk
 
S
, et al. 
Identification of acute myeloid leukaemia associated microRNA expression patterns.
Br J Haematol
2008
, vol. 
140
 
2
(pg. 
153
-
161
)
87
Lutherborrow
 
M
Bryant
 
A
Jayaswal
 
V
, et al. 
Expression profiling of cytogenetically normal AML identifies microRNAs that target genes involved in monocytic differentiation.
Am J Hematol
2011
, vol. 
86
 
1
(pg. 
2
-
11
)
88
Marcucci
 
G
Radmacher
 
MD
Maharry
 
K
, et al. 
MicroRNA expression in cytogenetically normal acute myeloid leukemia.
N Engl J Med
2008
, vol. 
358
 
18
(pg. 
1919
-
1928
)
89
Mariathasan
 
S
Monack
 
DM
Inflammasome adaptors and sensors: intracellular regulators of infection and inflammation.
Nat Rev Immunol
2007
, vol. 
7
 
1
(pg. 
31
-
40
)
90
Schwind
 
S
Maharry
 
K
Radmacher
 
MD
, et al. 
Prognostic significance of expression of a single microRNA, miR-181a, in cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study.
J Clin Oncol
2010
, vol. 
28
 
36
(pg. 
5257
-
5264
)
91
O'Connell
 
RM
Chaudhuri
 
AA
Rao
 
DS
Baltimore
 
D
Inositol phosphatase SHIP1 is a primary target of miR-155.
Proc Natl Acad Sci U S A
2009
, vol. 
106
 
17
(pg. 
7113
-
7118
)
92
Costinean
 
S
Sandhu
 
SK
Pedersen
 
IM
, et al. 
Src homology 2 domain-containing inositol-5-phosphatase and CCAAT enhancer-binding protein beta are targeted by miR-155 in B cells of Emu-MiR-155 transgenic mice.
Blood
2009
, vol. 
114
 
7
(pg. 
1374
-
1382
)
93
Georgantas
 
RW
Hildreth
 
R
Morisot
 
S
, et al. 
CD34+ hematopoietic stem-progenitor cell microRNA expression and function: a circuit diagram of differentiation control.
Proc Natl Acad Sci U S A
2007
, vol. 
104
 
8
(pg. 
2750
-
2755
)
94
Fazi
 
F
Racanicchi
 
S
Zardo
 
G
, et al. 
Epigenetic silencing of the myelopoiesis regulator microRNA-223 by the AML1/ETO oncoprotein.
Cancer Cell
2007
, vol. 
12
 
5
(pg. 
457
-
466
)
95
Johnnidis
 
JB
Harris
 
MH
Wheeler
 
RT
, et al. 
Regulation of progenitor cell proliferation and granulocyte function by microRNA-223.
Nature
2008
, vol. 
451
 
7182
(pg. 
1125
-
1129
)
96
Eyholzer
 
M
Schmid
 
S
Wilkens
 
L
Mueller
 
BU
Pabst
 
T
The tumour-suppressive miR-29a/b1 cluster is regulated by CEBPA and blocked in human AML.
Br J Cancer
2010
, vol. 
103
 
2
(pg. 
275
-
284
)
97
Garzon
 
R
Heaphy
 
CEA
Havelange
 
V
, et al. 
MicroRNA 29b functions in acute myeloid leukemia.
Blood
2009
, vol. 
114
 
26
(pg. 
5331
-
5341
)
98
Mott
 
JL
Kobayashi
 
S
Bronk
 
SF
Gores
 
GJ
Mir-29 regulates Mcl-1 protein expression and apoptosis.
Oncogene
2007
, vol. 
26
 
42
(pg. 
6133
-
6140
)
99
Baltimore
 
D
Boldin
 
MP
O'Connell
 
RM
Rao
 
DS
Taganov
 
KD
MicroRNAs: new regulators of immune cell development and function.
Nat Immunol
2008
, vol. 
9
 
8
(pg. 
839
-
845
)
100
Helgason
 
CD
Kalberer
 
CP
Damen
 
JE
, et al. 
A dual role for Src homology 2 domain-containing inositol-5-phosphatase (SHIP) in immunity: aberrant development and enhanced function of B lymphocytes in Ship−/− mice.
J Exp Med
2000
, vol. 
191
 
5
(pg. 
781
-
794
)
101
Helgason
 
CD
Damen
 
JE
Rosten
 
P
, et al. 
Targeted disruption of SHIP leads to hemopoietic perturbations, lung pathology, and a shortened life span.
Genes Dev
1998
, vol. 
12
 
11
(pg. 
1610
-
1620
)
102
Luo
 
J-M
Yoshida
 
H
Komura
 
S
, et al. 
Possible dominant-negative mutation of the SHIP gene in acute myeloid leukemia.
Leukemia
2003
, vol. 
17
 
1
(pg. 
1
-
8
)
103
Hirai
 
H
Zhang
 
P
Dayaram
 
T
, et al. 
C/EBPβ is required for ‘emergency’ granulopoiesis.
Nat Immunol
2006
, vol. 
7
 
7
(pg. 
732
-
739
)
104
Krivtsov
 
AV
Twomey
 
D
Feng
 
Z
, et al. 
Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9.
Nature
2006
, vol. 
442
 
7104
(pg. 
818
-
822
)
105
Calin
 
GA
Ferracin
 
M
Cimmino
 
A
, et al. 
A microRNA signature associated with prognosis and progression in chronic lymphocytic leukemia.
N Engl J Med
2005
, vol. 
353
 
17
(pg. 
1793
-
1801
)
106
Yanaihara
 
N
Caplen
 
N
Bowman
 
E
, et al. 
Unique microRNA molecular profiles in lung cancer diagnosis and prognosis.
Cancer Cell
2006
, vol. 
9
 
3
(pg. 
189
-
198
)
107
Iorio
 
MV
Ferracin
 
M
Liu
 
C-G
, et al. 
MicroRNA gene expression deregulation in human breast cancer.
Cancer Res
2005
, vol. 
65
 
16
(pg. 
7065
-
7070
)
108
Han
 
Y-C
Park
 
CY
Bhagat
 
G
, et al. 
microRNA-29a induces aberrant self-renewal capacity in hematopoietic progenitors, biased myeloid development, and acute myeloid leukemia.
J Exp Med
2010
, vol. 
207
 
3
(pg. 
475
-
489
)
109
Blum
 
W
Garzon
 
R
Klisovic
 
RB
, et al. 
Clinical response and miR-29b predictive significance in older AML patients treated with a 10-day schedule of decitabine.
Proc Natl Acad Sci U S A
2010
, vol. 
107
 
16
(pg. 
7473
-
7478
)
110
Goff
 
LA
Davila
 
J
Swerdel
 
MR
, et al. 
Ago2 immunoprecipitation identifies predicted microRNAs in human embryonic stem cells and neural precursors.
PLoS ONE
2009
, vol. 
4
 
9
pg. 
e7192
 
111
Pant
 
BD
Musialak-Lange
 
M
Nuc
 
P
, et al. 
Identification of nutrient-responsive Arabidopsis and rapeseed microRNAs by comprehensive real-time PCR profiling and small RNA sequencing.
Plant Physiol
2009
, vol. 
150
 
3
(pg. 
1541
-
1555
)
112
Jima
 
DD
Zhang
 
J
Jacobs
 
C
, et al. 
Deep sequencing of the small RNA transcriptome of normal and malignant human B cells identifies hundreds of novel microRNAs.
Blood
2010
, vol. 
116
 
23
(pg. 
e118
-
e127
)
113
Yang
 
M
Mattes
 
J
Discovery, biology and therapeutic potential of RNA interference, microRNA and antagomirs.
Pharmacol Ther
2008
, vol. 
117
 
1
(pg. 
94
-
104
)
114
Garzon
 
R
Marcucci
 
G
Croce
 
CM
Targeting microRNAs in cancer: rationale, strategies and challenges.
Nat Rev Drug Discov
2010
, vol. 
9
 
10
(pg. 
775
-
789
)
115
Krützfeldt
 
J
Rajewsky
 
N
Braich
 
R
, et al. 
Silencing of microRNAs in vivo with ‘antagomirs.’
Nature
2005
, vol. 
438
 
7068
(pg. 
685
-
689
)
116
Elmén
 
J
Lindow
 
M
Schütz
 
S
, et al. 
LNA-mediated microRNA silencing in non-human primates.
Nature
2008
, vol. 
452
 
7189
(pg. 
896
-
899
)

Author notes

*

G.M and K.M. contributed equally to this study.

Sign in via your Institution