Molecular dysfunctions in acute myeloid leukemia revealed by integrated analysis of microRNA and transcription factor

  • Authors:
    • Xiao-Cong Lin
    • Yong Xu
    • Guo-Ping Sun
    • Jin-Li Wen
    • Ning Li
    • Yu-Ming Zhang
    • Zhi-Gang Yang
    • Hai-Tao Zhang
    • Yong Dai
  • View Affiliations

  • Published online on: April 15, 2016     https://doi.org/10.3892/ijo.2016.3489
  • Pages: 2367-2380
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Acute myeloid leukemia (AML) is a heterogenic hematological malignancy with pathogenesis that has yet to be elucidated. MicroRNAs (miRNAs) and transcription factors (TFs) are two major regulators of gene expression, which may play important roles in the etiology of AML. However, the global regulation of gene expression in AML, involving miRNAs and TFs, still remains elusive. To characterize the global role of miRNAs and TFs in AML pathogenesis, large scale expression profiling of miRNA and TF was performed using miRNA sequencing and TF array technology, respectively, and validated by qPCR. In the present study, 308 miRNAs and 84 TFs were identified to be differentially expressed (fold-change ≥2.0) in AML samples relative to their controls. After integrating the expression profiling data into bioinformatic analysis, we identified 1,462 miRNA-gene pairs, 982 TF-gene pairs and 296 TF-miRNA pairs. By merging these regulatory relations together, we constructed a comprehensive AML-specific miRNA-TF regulatory network. In this network, we identified 22 hub miRNAs and 11 hub TFs. KEGG pathway analysis showed that the network nodes were significantly enriched in 33 different pathways, of which the AML pathway was the most significant. After analyzing the topology of the subnetwork, we propose that TCF3 was a potential key regulator in this regulatory network. In conclusion, this is the first study perform on global expression profiling of miRNAs and TFs relating to AML. These results may enhance our understanding of the molecular mechanisms underlying AML and provide potential targets for future therapeutics.

Introduction

Acute myeloid leukemia (AML) is an aggressive hematological malignancy accounting for ~80% of all adult leukemia cases, and encompasses 15–20% of leukemia cases in children (1,2). It is estimated that nearly 15% of mortality among patients with hematologic malignancy were attributed to AML (3), and more than 13,000 new cases were diagnosed each year in the USA (4), thus, making AML a major cause of hematologic disorders. AML is usually characterized by malignant clonal proliferation of hematopoietic stem cells and progenitors (blasts) in the bone marrow with the complete or partial blockage at different stages of myeloid differentiation (1,2). Despite advances in chemotherapy and hematopoietic stem cell transplantation, the outcome of AML patients have not improved substantially in the last four decades, with an overall 5-year survival rate of ~25% (1,2,5). Relapse occurs frequently, as ~50–70% of AML patients who achieve complete remission within three years after frontline therapy (2). Among these relapsed patients, only one-third are salvageable with current treatment regimens (5). Therefore, it is urgently needed to search for new therapeutic targets for AML. Securing further advances in therapy is dependent on our increasing understanding of the factors and molecular mechanisms of leukemogenesis in AML. Currently, insight into the pathogenesis of AML has largely come from the investigations of cytogenetic abnormalities and molecular genetic mutations. Many chromosomal structural aberrations and somatic mutations have been identified, which include rearrangements of RUNX1-RNNX1T1, CBFB-MYH11, PML-RARA and MLL gene (6), mutations of CEBPA, NPM1, FLT3, DNMT3A, TET2, IDH1/2 and ASXL1 gene (1,7,8). Despite the pathological role and prognostic implications of these genes have been well elucidated, understanding of the multistep pathogenesis of AML remains limited. Specifically, the regulatory networks of AML gene expression are still unclear.

miRNAs are small non-protein coding RNA molecules of ~19–25 nucleotides that mediate post-transcriptional regulation of gene expression level (9). Pri-miRNAs are transcribed by RNA polymerase II and subsequently processed by the Drosha-DGCR8 enzyme complex in the nucleus to form ~70 nt hairpin pre-miRNAs. Finally, pre-miRNAs were cleaved in the cytoplasm by the endoribonuclease Dicer to yield mature miRNAs. The mature miRNA may bind to the 3′-UTR of target mRNAs through its seed region, which leads to mRNA degradation or inhibition of translation (10). miRNAs play important roles in a variety of biological processes and are implicated in the initiation and progression of many types of human cancers, including AML (11,12). It has been reported that ectopic expression of miR-29a would induce hematopoietic stem cells/progenitors progresses to AML in a mouse model (13). High expression of miR-100 and miR-375 was found in pediatric AML patients, which were correlated with poorer relapse-free and overall survival (14,15). miR-193a expression is downregulated in AML1/ETO-positive leukemia cells, suppression of miR-193a expands the oncogenic activity of the fusion protein AML1-ETO involving a feedback circuitry in miR-193a and AML1-ETO/DNMTs/HDACs (16). Recently, increased miR-181a expression was shown to be associated with improved prognosis in cytogenetically normal AML. In xenograft mouse models of AML, ectopic miR-181a expression inhibits tumor growth (17). Although the aberrant expression and molecular function of AML-related miRNAs were reported in these investigations, the global miRNA regulatory network in AML remains unknown.

TFs are trans-acting protein factors that control the transcription of target genes through specifically binding to the TFBS located within promoter region of target genes (18). As two major types of regulators of gene expression, TFs and miRNAs are able to tightly coordinate to ensure precise and accurate gene expression. TF and miRNA may reciprocally regulate one another and both can regulate the expression of target genes in a combinatorial manner (18,19). It has been reported that miR-223 and transcription factor E2F1 regulate each other to form negative feedback loop in AML (20). Furthermore, transcription factor C/EBPα can exert its effects by inducing miR-30c inactivating the Notch1 protein and enhancing granulocytic differentiation in AML (21). Nevertheless, global regulation of gene expression involved in the TFs and miRNAs in AML are still poorly understood. Therefore, integrated analysis correlating changes in the expression patterns of miRNAs and TFs, as it relates to AML pathogenesis, requires examination.

In the present study, we analyzed miRNA and TF expression profiles in bone marrow samples of AML patients and control groups, utilizing miRNA sequencing and TF array technology, respectively. Based on a combined strategy of the target prediction of miRNAs and TFs, we integrated differentially expressed miRNAs, AML candidate genes and differentially expressed TFs to construct a comprehensive miRNA-TF mediated regulatory network specifically for AML. GO and pathway analysis was performed to determine associated functions and signaling pathway of the network nodes. After calculating the number of the node degree, we found some hub miRNAs and TFs, and further investigated their regulation in the subnetwork. Our results demonstrate that the altered expression levels of miRNAs and TFs may have implications in AML pathogenesis, and that integrative analysis of miRNAs and TFs may provide a new foundation for molecularly targeted therapy of AML in the future.

Materials and methods

Patient samples and ethics statement

A total of 25 bone marrow biopsy samples, including 15 patients with newly diagnosed and untreated AML and 10 age-matched controls, were enrolled in the study. The AML samples were collected at Shenzhen People's Hospital (Shenzhen, China) before any chemo- or radiotherapeutic treatment. Diagnosis and classification of AML were determined according to the French-American-British classification system (22,23). To serve as controls, the samples with normal bone marrow morphology were provided from 10 patients with unexplained anemia or fever by the Affiliated Hospital of Guangdong Medical University (Zhanjiang, China). All the clinical characteristics of AML patients and controls are shown in Table I. The study was carried out according to the Helsinki convention criteria and approved by the ethics committee of the Shenzhen People's Hospital and the Affiliated Hospital of Guangdong Medical University. Written informed consent to use biological samples and clinical data was obtained from patients or their parents.

Table I

Clinical characteristics of AML patients and controls.

Table I

Clinical characteristics of AML patients and controls.

CharacteristicsAML patients (n=15)Control patients (n=10)
Age (years)18–77 (mean 40)10–73 (mean 39)
Gender (n)
 Male96
 Female64
FAB subtypes (n)
 M01-
 M13-
 M25-
 M32-
 M42-
 M51-
 M61-
Leukemic blasts in bone marrow (%)30–99 (mean 78)-
Disease (n)
 AML15-
 Unexplained anemia-9
 Unexplained fever-1
Total RNA extraction

Total RNA was extracted using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) and a RNeasy kit (Qiagen, Hilden, Germany) following manufacturer's instructions, to include a DNase digestion step. RNA purity and concentration were determined using a NanoDrop ND-1000 spectrophotometer (NanoDrop Technologies, Wilmington, DE, USA) measuring absorbance at 230, 260 and 280 nm.

miRNA sequencing and data analysis

Total RNA was ligated sequentially to 3′ and 5′ RNA adapters using T4 RNA ligase (Promega, Madison, WI, USA). Ligation products were then reverse-transcribed and PCR amplified using Illumina's proprietary RT primers and amplification primers (Illumina, San Diego, CA, USA). Subsequently, ~135–155 bp PCR amplified fragments (correspond to ~15–35 nt small RNAs) were isolated and purified from the PAGE gels. After quantified with Agilent 2100 Bioanalyzer (Agilent Technologies, Palo Alto, CA, USA), the completed libraries were diluted to a final concentration of 8 pM and loaded on the Illumina cBot for cluster generation using TruSeq Rapid SR cluster kit (Illumina) according to the manufacturer's protocol. Finally, the sequencing was carried out on Illumina HiSeq 2000 using TruSeq Rapid SBS kit (Illumina) as recommended by the manufacturer.

After sequencing, image analysis and base calling were carried out using Off-Line Basecaller software (version V1.8.0; Illumina). The low quality reads were removed from raw sequencing reads by Solexa CHASTITY quality control filter. Subsequently, 3′ adapter sequences were deleted from clean reads and the reads that were shorter than 15 nt were excluded from further analysis. To identify miRNAs, the 3′-adapter-trimmed-reads were aligned to known human pre-miRNAs in the miRBase database (Release 19.0, http://www.mirbase.org/) using Novoalign software (Version v2.07.11, http://www.novocraft.com/) with at most one mismatch. In order to correct for the difference in read counts between samples, the read counts per miRNA in each sample were normalized to tpm (the clone number of transcripts per million based on the sum number of reads aligned to known human pre-miRNAs in miRBase 19.0). Differentially expressed miRNAs may be identified by a fold-change filtering. The threshold is fold-change ≥2.0 (Based on the normalized most abundant tag counts).

Transcription factor/DNA array and computational analysis

The transcription factor/DNA array analysis was carried out using TranSignal™ Protein/DNA Combo Arrays (Panomics, Redwood City, CA, USA) following the protocols provided by the manufacturer, which included 345 major transcription factors. Briefly, nuclear proteins were extracted using the Panomics nuclear extract kit (Panomics) and protein concentrations were determined by BCA protein assay kit (KangChen Bio-Tech Inc., Shanghai, China). Nuclear extract (10 μg) were incubated with 10 μl of biotin-labelled DNA binding oligonucleotides (TranSignal™ Probe Mix) for 30 min at 15°C to allow the formation of transcription factor/DNA complexes. Such complexes were isolated from the free probes by spin column separation system (Panomics). The bound DNA probes were extracted, denatured and hybridized to oligonucleotides (representing 345 consensus binding sequences for TFs) on TransSignal array membrane at 42°C overnight. Subsequently, the blots were washed and incubated with HRP and HRP substrate working solution (Millipore, Billerica, MA, USA), and exposed to Hyperfilm ECL (Amersham Pharmacia Biotech, Uppsala, Sweden). Hybridization signals were detected using GBoX Imaging System (Syngene, Cambridge, UK), and quantitative analysis of the resulting spots were performed using the ScanAlyze software (version 1.0.3, http://graphics.stanford.edu/software/scanalyze/). Any spot showing at least 2-fold increase or decrease is considered significant.

Quantitative PCR

Total RNA from each sample was reverse transcribed to generate cDNA with gene-specific primer (for miRNAs) and oligo dT primer (for TFs) using a Superscript™ III reverse transcriptase kit (Invitrogen, Carlsbad, CA, USA) in accordance with the manufacturer's instructions. QPCR was carried out in a total reaction volume of 10 μl, including 2 μl of template cDNA, 5 μl of 2X SYBR-Green PCR Master Mix (Applied Biosystems, Foster City, CA, USA), 0.5 μl of PCR forward primer (10 μM), 0.5 μl of PCR reverse primer (10 μM) and 2 μl of double-distilled water. The reactions were incubated at 95°C for 10 min, followed by 40 cycles at 95°C for 10 sec, 60°C for 60 sec. All reactions were run in triplicate. After PCR amplification, melt curve analysis was carried out to determine the reaction specificity. Human U6 snRNA and 18S rRNA was used to normalize the expression levels of miRNAs and TFs, respectively. Expression fold-changes were calculated using the 2−ΔΔCt method (24). The differences in gene expression levels between AML samples and controls were compared using the Student's t-test. Statistical significance was set at P<0.05. All primers used in cDNA synthesis and qPCR are shown in Tables II, III and IV.

Table II

Primers used for cDNA synthesis of miRNAs.

Table II

Primers used for cDNA synthesis of miRNAs.

miRNAsPrimer for cDNA synthesis (5′-3′)
miR-9-5p GTCGTATCCAGTGCGTGTCGTGGAGTCGGCAATTGCACTGGATACGACTCATACA
miR-155-5p GTCGTATCCAGTGCGTGTCGTGGAGTCGGCAATTGCACTGGATACGACACCCCTA
miR-100-5p GTCGTATCCAGTGCGTGTCGTGGAGTCGGCAATTGCACTGGATACGACCACAAG
miR-223-3p GTCGTATCCAGTGCGTGTCGTGGAGTCGGCAATTGCACTGGATACGACTGGGGTA
miR-16-5p GTCGTATCCAGTGCGTGTCGTGGAGTCGGCAATTGCACTGGATACGACCGCCAAT
miR-106b-5p GTCGTATCCAGTGCGTGTCGTGGAGTCGGCAATTGCACTGGATACGACATCTGCA
U6 snRNA CGCTTCACGAATTTGCGTGTCAT

Table III

Primers used for real-time quantitative PCR of miRNAs.

Table III

Primers used for real-time quantitative PCR of miRNAs.

miRNAsSense primer (5′-3′)Antisense primer (5′-3′)Product (bp)
miR-9-5p GGGGGTCTTTGGTTATCTA CAGTGCGTGTCGTGGA68
miR-155-5p GGGGTAATGCTAATCGTGA CAGTGCGTGTCGTGGAG66
miR-100-5p GCAACCCGTAGATCCGAA CAGTGCGTGTCGTGGAGT62
miR-223-3p GGGGTGTCAGTTTGTCAAA CAGTGCGTGTCGTGGAGT66
miR-16-5p GGGTAGCAGCACGTAAATA CAGTGCGTGTCGTGGAGT65
miR-106b-5p GGGGGTAAAGTGCTGACAGT GTGCGTGTCGTGGAGTCG64
U6 snRNA GCTTCGGCAGCACATATACTAAAAT CGCTTCACGAATTTGCGTGTCAT89

Table IV

Primers used for real-time quantitative PCR of TFs.

Table IV

Primers used for real-time quantitative PCR of TFs.

TFsSense primer (5′-3′)Antisense primer (5′-3′)Product (bp)
MYC ACACATCAGCACAACTACGC CCTCTTGACATTCTCCTCGGT159
NFKB1 ACTGGCTGAGCGGATGCATC TGCTGTGGTCAGAAGGAATG165
NR2F1 ATCGAGAGCCTGCAGGAGAA CTACCAAACGGACGAAGAAGAG163
FOXO1 GCAACTACAGCCAAAATCAC TCAGAGAGCTACCAAGGATTC152
FOXL1 TGAGGTTTGATGGCAGGAAT GATTTTCGTTGCAGACCTCTTC171
TFAP2A TTGGGTACGTGTGCGAAA TCTGTTTTGTAGCCAGGAGCAT120
18S rRNA CCTGGATACCGCAGCTAGGA GCGGCGCAATACGAATGCCCC112
Association analysis of the different expression of miRNAs and TFs

Target gene prediction for different expression miRNAs. Differentially expressed miRNAs (fold change, ≥2.0) were subjected to bioinformatic analysis for target gene prediction mainly by combinatorial utilization of four different online databases, including miRanda (version v5, http://www.ebi.ac.uk/enright-srv/microcosm/htdocs/targets/v5/), TargetScan (Release 6.2, http://www.targetscan.org/), PicTar (Release 2007, http://pictar.mdc-berlin.de/cgi-bin/PicTar_vertebrate.cgi) and miRTarBase (Release 4.5, http://mirtarbase.mbc.nctu.edu.tw/) (25,26). To decrease the number of false-positive results, we integrated the predicted targets from miRanda, TargetScan and PicTar based on sequence complementarity, evolutionary conservation and free energy of RNA duplexes, which predicted by at least two of three databases were accepted as positive (25). (TFs were treated as genes when predicting miRNA→TF regulatory relations). Subsequently, we merged the results with experimentally validated miRNA targets from miRTarBase.

Combined AML candidate genes with miRNA target prediction

To avoid the redundancy, AML-associated genes were obtained from the MalaCards database (version 1.08.564, http://www.malacards.org/, MalaCards ID: LKM061) (27) and overlapped with the predicted miRNA targets above to form a miRNA→gene regulatory relations. The extracted miRNA and target gene pairs were then subjected to transcription factor binding site (TFBS) analysis.

TFBS prediction for different expression miRNAs and its pair target genes

To retrieve predicted TFBS information, We utilized the data underlying the TFBS Conserved Track (http://genome.ucsc.edu/cgi-bin/hgTables?hgsid=350051003&hgta_doSchemaDb=hg19&hgta_doSchemaTable=tfbsConsFactors) at the UCSC Genome Browser (28). These binding sites are conserved across the human/mouse/rat alignment. To further reduce the false-positive prediction, P-value of 0.05 was used as a cut-off for high-quality TFBSs. A gene is identified as the target of a TF if at least one TFBS is located in the gene's promoter region (5,000 bp upstream and 1,000 bp downstream of the TSS) and its P-value was <0.05. miRNA overlaps with a known host transcript (mRNA/lncRNA) and serve as a part of the same transcription unit. Therefore, the promoter for this specific transcript is used as the miRNA promoter. We identified the target miRNAs of the TFs by using the same approach as for protein-coding genes and lncRNAs. A miRNA is regarded as the target of a TF if at least one TFBS falls within the TSS region (from 5,000 bp upstream to 1,000 bp downstream) of the host gene or lncRNA and its P-value was <0.05.

Combined differentially expressed TFs with TFBS prediction

To further increase the accuracy of TFBS prediction, differentially expressed TFs (fold-change, ≥2.0) were overlapped with the predicted TFs above. Subsequently, we extracted TFs and miRNA pairs, TFs and miRNA target genes pairs to form TF→miRNA and TF→gene regulatory relations, respectively.

miRNA-TF regulatory network construction and network node analysis

The TF→miRNA and TF→gene interactions were incorporated into miRNA→gene interactions to construct a comprehensive miRNA-TF mediated regulatory network, which was visualized using Gephi software (version 0.8.1-beta, http://gephi.github.io/). For further network functional evaluation, GO analysis and KEGG pathway analysis were performed using the Functional Annotation Tool of DAVID database (version v6.7, http://david.abcc.ncifcrf.gov/) (29) to identify the functional categories enriched and pathways for the network nodes. GO term enrichment and the KEGG pathways utilized significant P-values (<0.05) relating to the nodes of miRNA-TF regulatory network. To assess network characteristics, the node degree is measured by the number of direct links of the node in the network. Nodes having a total degree ≥20 were defined as hub nodes inside the network. The subnetworks were then constructed by combining all the directly linked nodes for the hubs.

Statistical analysis

Statistical analyses were performed are presented as means ± standard deviation (SD). The using SPSS for Windows (version 13.0; SPSS, Inc.). All values statistical difference between means were analyzed by the Student's t-test and P-value <0.05 was considered significant.

Results

Aberrant miRNA and TF expression in AML

To obtain AML-related miRNAs and TFs, we analyzed miRNA and TF expression profiles in bone marrow samples of AML patients and control groups, using miRNA sequencing and TF array technology, respectively. Among the miRNA transcripts examined, we found 308 miRNAs were differentially expressed (fold-change ≥2.0) in AML samples relative to their controls (Fig. 1A), with 233 being upregulated, while 75 were downregulated. miR-206 (fold-change, 1144) was the most significantly upregulated miRNA, while miR-941 (fold-change, 184.2) was the most significantly downregulated miRNA.

The TF expression profiling data showed 84 TFs to be differentially expressed (fold-change ≥2.0) in AML samples relative to their controls (Fig. 1B), with 76 upregulated, while 8 were downregulated. Among these TFs, PREB (fold-change, 25.9) showed the highest degree of upregulation, while HiNF-B (fold-change, 2.8) was the most downregulated TF.

Quantitative PCR validation

To validate the miRNA sequencing and TF array results, 6 differentially expressed miRNAs and 6 differentially expressed TFs were selected and analyzed via qPCR. For the miRNAs, the results demonstrated that miR-155-5p, miR-9-5p and miR-100-5p were upregulated and that miR-223-3p, miR-16-5p and miR-106b-5p were down-regulated in the AML samples compared with control samples (P<0.05 for each miRNAs; Fig. 2A). For the TFs, the expression of MYC, NFKB1, NR2F1, FOXO1, FOXL1 and TFAP2A showed statistically significant differences between the two sets of bone marrow samples (all P<0.05; Fig. 2B). These qPCR results were consistent with the miRNA sequencing and TF array data.

miRNA and TF regulatory network in AML

The basic integrated regulatory network for miRNA and TF consists of three types of interactions: miRNA→gene (including miRNA→TF), TF→miRNA and TF→gene regulatory relations. After removing a few isolated nodes, we merged the three regulatory relations and constructed a miRNA-TF regulatory network for AML (Fig. 3). The numbers of nodes and pairs in the network are listed in Table V. Among the AML-related miRNAs in the network, most of (117/173) them are upregulated. Moreover, there were three pairs of miRNA→gene regulatory relationships (miR-126-5p and miR-126-3p represses PLK2 and miR-17-5p represses RUNX1) that have been experimentally confirmed in AML (30,31). Two TFs (NFKB1 and MYC) in this network have been reportedly associated with the development of AML (32,33). NFKB1 and MYC transcriptionally suppressed miR-29b-3p expression by binding to its promoter, and MYC was confirmed as a transcriptional repressor of miR-15a-5p, which were in accord with the same TF→miRNA regulatory relationships in our network (34,35). As key regulators of gene expression, miRNAs and TFs may reciprocally regulate each other to form FBLs, or co-regulate the expression of the same targets to form FFLs (18). In our miRNA-TF regulatory network, we identified 13 FBL and 1156 FFL motifs, reflecting the tight relationships between miRNAs and TFs in the network.

Table V

Summary of relationships in the AML-related miRNA and transcription factor (TF) regulatory network.

Table V

Summary of relationships in the AML-related miRNA and transcription factor (TF) regulatory network.

RelationshipNo. of pairsNo. of miRNAsNo. of genesNo. of TFs.
miRNA-genea1462173150-
miRNA-TFb6449-5
TF-genec982-26410
TF-miRNAd296114-11

a miRNA repression of gene expression.

b miRNA repression of TF expression.

c TF regulation of gene expression.

d TF regulation of miRNA expression.

Biological functions of the miRNA-TF regulatory network

To investigate the functional groupings of the miRNA-TF regulatory network in AML, we applied the DAVID database to classify the distinct network nodes according to their GO molecular function, cellular component and biological process terms (Fig. 4). In the GO molecular function classification, a large proportion of nodes associated with binding, protein binding and nucleic acid binding (Fig. 4A). The nodes related to cellular components were mainly located in the cell parts, cell and intracellular (Fig. 4B). The GO biological processes classification showed that nodes were mainly involved in cellular process, metabolic process and biological regulation (Fig. 4C). To ascertain which pathways are enriched in the network nodes, KEGG pathway analysis was also performed by using the DAVID database. The results indicate that the nodes were significantly enriched in 33 different pathways, of which the AML pathway was the most significant followed by the pathways in cancer and Jak-STAT signaling pathways (Table VI). Among these pathways, 17 were related to hematopoiesis and leukemia. In addition, AML pathway, apoptosis pathway, Jak-STAT signaling pathway, MAPK signaling pathway, Wnt signaling pathway, p53 signaling pathway, cell cycle pathway and ABC transporter pathway have been previously experimentally verified to be involved in AML (2,3642).

Table VI

Pathway analysis for network nodes in AML-related miRNA and transcription factor regulatory network.

Table VI

Pathway analysis for network nodes in AML-related miRNA and transcription factor regulatory network.

Pathway IDDefinitionGene count%Fisher-P-value
hsa05221Acute myeloid leukemia249.091.31E-21
hsa05200Pathways in cancer4316.295.77E-18
hsa04630Jak-STAT signaling pathway228.331.21E-09
hsa05220Chronic myeloid leukemia155.681.32E-08
hsa05215Prostate cancer166.061.67E-08
hsa04640Hematopoietic cell lineage155.688.18E-08
hsa04210Apoptosis124.552.65E-05
hsa05210Colorectal cancer114.171.05E-04
hsa04060Cytokine-cytokine receptor interaction197.203.44E-04
hsa05222Small cell lung cancer103.795.23E-04
hsa05213Endometrial cancer83.035.57E-04
hsa04012ErbB signaling pathway103.796.78E-04
hsa05211Renal cell carcinoma93.416.95E-04
hsa04660T cell receptor signaling pathway114.178.30E-04
hsa05212Pancreatic cancer93.418.41E-04
hsa05219Bladder cancer72.650.001013
hsa05216Thyroid cancer62.270.001129
hsa02010ABC transporters72.650.001302
hsa04722Neurotrophin signaling pathway114.170.002384
hsa04010MAPK signaling pathway176.440.003123
hsa04310Wnt signaling pathway124.540.003263
hsa05218Melanoma83.030.00355
hsa04910Insulin signaling pathway114.170.004425
hsa04662B cell receptor signaling pathway83.030.004828
hsa04520Adherens junction83.030.005583
hsa04916Melanogenesis93.410.006365
hsa04110Cell cycle103.790.008204
hsa04672Intestinal immune network for IgA production62.270.01157
hsa04650Natural killer cell mediated cytotoxicity103.790.012086
hsa04664Fc epsilon RI signaling pathway72.650.021766
hsa04620Toll-like receptor signaling pathway83.030.022915
hsa05214Glioma62.270.031252
hsa04115p53 signaling pathway62.270.04151
Network hubs and subnetworks for hubs in the miRNA-TF regulatory network

To ascertain which nodes potentially have the most influence on the overall behavior of the networks, we identified the hub nodes based on the highest total degrees inside the network. As shown in Tables VII and VIII, 22 miRNAs and 11 TFs were defined as hub nodes, which having a total degree ≥20. It is of interest that five of the 22 hub miRNAs belonged to the miR-15 family, namely miR-15a-5p/miR-15b-5p/miR-16-5p/miR-424-5p/miR-195-5p (Table VII), further indicating the important role of the miR-15 family in the regulatory network of AML. Notably, more than half of the 11 hub TFs were either notorious leukemia regulators, such as NFE2 (43) and MYC (33), or related to leukemia development and progression, such as NFKB1 (32), TCF3 (44), MAX (45) and FOXO1 (46). These findings are a preliminary reflection on the robustness of our network. Among these hub nodes, two hub miRNAs (miR-15a-5p and miR-125b-5p) and two hub TFs (MYC and NFKB1) have been previously implicated in AML (12,32,33,47). To further investigate the regulation of the four hub nodes, we extracted their subnetworks by including all their directly linked nodes in the miRNA-TF regulatory network (Fig. 5). The subnetwork analysis indicated that many pathways related to AML and other cancers (Table IX), which suggested that miR-15a-5p, miR-125b-5p, MYC and NFKB1 were important in cancer development including AML. In addition, TCF3 stood out as a promising regulator and gene, which links to all the four subnetworks. As a TF, it regulates miR-125b-5p (Fig. 5B) and MYC (Fig. 5C); while in its capacity as a gene, it is regulated by miR-15a-5p (Fig. 5A), MYC (Fig. 5C) and NFKB1 (Fig. 5D). In particular, 17 predicted target genes (CEBPA, FLT3, STAT5A, RELA, STAT5B, PIM1, SPI1, RUNX1T1, LEF1, PIM2, KIT, JUP, NRAS, EIF4EBP1, KRAS, RARA and MYC) of TCF3 in the four subnetworks participated in the AML pathway. These analyses suggest that TCF3 may play an important role in the miRNA-TF regulatory network linked to AML.

Table VII

Hub miRNAs in AML-related miRNA and transcription factor regulatory network.

Table VII

Hub miRNAs in AML-related miRNA and transcription factor regulatory network.

TopmiRNAIn-degreeOut-degreeTotal degree
1miR-335-5p14142
2miR-124-3p04141
3miR-16-5p43034
4miR-30a-5p42630
5miR-26b-5p12829
6miR-23b-3p02828
7miR-15a-5p62127
8miR-23a-3p02424
9miR-338-5p32124
10miR-30c-5p42024
11miR-15b-5p41923
12miR-17-5p71623
13miR-181b-5p41923
14miR-20a-5p71522
15miR-144-3p02121
16miR-192-5p31821
17miR-424-5p51520
18let-7e-5p02020
19miR-125b-5p31720
20miR-186-5p11920
21miR-195-5p31720
22miR-9-5p11920

Table VIII

Hub transcription factors in AML-related miRNA and transcription factor regulatory network.

Table VIII

Hub transcription factors in AML-related miRNA and transcription factor regulatory network.

TopTranscription factorIn-degreeOut-degreeTotal degree
1TCF314192206
2MYC9186195
3MEF2A0167167
4NFKB18153161
5MAX0152152
6FOXO14398141
7NFKB238083
8NFE207373
9NR2F107373
10NKX2-207171
11FOXL105959

Table IX

Pathways enriched among the nodes of four subnetworks.

Table IX

Pathways enriched among the nodes of four subnetworks.

SubnetworkEnriched pathway of subnetwork nodesaP-value
miR-15a-5pPathways in cancer2.66E-07
Acute myeloid leukemia3.25E-05
Small cell lung cancer1.40E-04
Prostate cancer1.75E-04
Cell cycle6.46E-04
MAPK signaling pathway0.001412
Colorectal cancer0.002502
Neurotrophin signaling pathway0.007495
Wnt signaling pathway0.012855
p53 signaling pathway0.021056
Chronic myeloid leukemia0.025301
Apoptosis0.033319
miR-125b-5pPathways in cancer0.001898
Colorectal cancer0.024629
Small cell lung cancer0.024629
Apoptosis0.026297
Prostate cancer0.027434
MYCAcute myeloid leukemia5.58E-13
Pathways in cancer3.73E-08
Prostate cancer4.59E-05
Jak-STAT signaling pathway2.55E-04
Thyroid cancer2.55E-04
Wnt signaling pathway0.001212
Chronic myeloid leukemia0.001297
Hematopoietic cell lineage0.002388
Endometrial cancer0.002413
Melanogenesis0.004401
Melanoma0.007421
MAPK signaling pathway0.008221
Bladder cancer0.010721
Cell cycle0.011628
Colorectal cancer0.013264
ErbB signaling pathway0.014936
Insulin signaling pathway0.015823
NFKB1Acute myeloid leukemia2.05E-12
Pathways in cancer8.66E-11
Jak-STAT signaling pathway2.73E-09
Chronic myeloid leukemia2.73E-06
T cell receptor signaling pathway4.17E-05
Prostate cancer6.79E-04
MAPK signaling pathway0.004636
Pancreatic cancer0.011110
B cell receptor signaling pathway0.012775
Neurotrophin signaling pathway0.016993
Small cell lung cancer0.018703
Hematopoietic cell lineage0.020218
NFKB1 Apoptosis0.021003
Toll-like receptor signaling pathway0.033987
Wnt signaling pathway0.036016
Cytokine-cytokine receptor interaction0.038854
Adipocytokine signaling pathway0.047655
Thyroid cancer0.048101
Epithelial cell signaling in Helicobacter pylori infection0.049434

a Pathways in italic are related to AML.

Discussion

AML is the most common form of aggressive leukemia in adults (1,2). Despite extensive efforts to elucidate the cytogenetic and molecular genetic mechanisms involved in disease occurrence and development, the pathogenesis of AML is still not fully understood due to the heterogeneity and complexity of this disease. The genomic complexity makes it difficult to identify hub regulators or genes in the pathogenesis of AML. By integrating diverse data sources, systems biology approaches provide a powerful tool for exploring the interactions and searching for key regulators between disease candidate genes on the network level, which may help to explain key aspects of disease pathogenesis and leading to new candidates for the putative therapeutic targets (48).

As major regulators of gene expression, miRNAs and TFs can function as tumor suppressors or oncogenes in a cooperative way to control gene expression, which triggering global alterations of genetic programs are involved in cell proliferation, differentiation, development and apoptosis in multiple human cancers (18). Ye et al (49) constructed a miRNA-TF regulatory network in T-cell acute lymphoblastic leukemia and demonstrated the roles of miR-19 (hub miRNA) and CYLD (hub gene) in the T-cell leukemogenesis. Therefore, understanding and applying regulatory network information for TF-miRNA-target genes could provide clues for key driver miRNAs and genes in human cancers and subsequently suggest novel therapeutic targets (50).

In the present study, we applied miRNA sequencing and TF array technology to quantitatively analyze the differential expression profiles of miRNAs and TFs, respectively, in bone marrow samples of AML patients and their age-matched controls (Fig. 1). The expression profiling data showed that 308 miRNAs and 84 TFs were differentially expressed (fold-change ≥2.0) in AML samples relative to their controls, with a subset of these findings further validated by qPCR (Fig. 2). It is difficult to achieve by only direct experimental methods to systematically infer the comprehensive miRNA-TF regulatory network, which has promoted development of computational approaches. Therefore, we integrated expression profiling data of miRNAs and TFs, computational miRNA and TF target prediction, and AML candidate genes obtained from MalaCards database to construct a miRNA and TF regulatory network specifically for AML (Fig. 3), which might provide important insights into the pathogenesis of AML. Moreover, we found that some miRNA→gene and TF→miRNA regulatory relationships in our network have been experimentally verified in previous studies. For further network functional evaluation, GO category and KEGG pathway annotation were applied to analyze the network node pool (Fig. 4 and Table VI). Among 33 enriched pathways, the AML pathway was the most significant (Table VI), more than half of these pathways were related to hematopoiesis and leukemia development, and eight pathways have been reported to be implicated in AML pathogenesis. These results preliminarily confirmed the reliability of our network.

In the miRNA-TF regulatory network, we identified 22 hub miRNAs and 11 hub TFs. With a large number of nodes linked to them, these hubs have the most influence on the networks overall behavior, which likely play critical regulatory roles in AML. To decipher this massive and complex network, and mine the key regulatory components, we subsequently constructed the subnetworks (Fig. 5) from two hub miRNAs (miR-15a-5p and miR-125b-5p) and two hub TFs (MYC and NFKB1) which were well-known regulators in AML. miR-15a-5p plays an important role in myeloid and erythroid differentiation, which was reported as a tumor suppressor in multiple myeloma, chronic lymphocytic leukemia and AML cells (51,52). Overexpression of miR-15a-5p significantly inhibited AML cell proliferation by downregulating the WT1 oncogene (51). miR-125b-5p is an oncogenic miRNA, which was highly expressed in AML carrying the t(2;11)(p21;q23) translocation and inhibited myeloid differentiation (12). Mice overexpressing miR-125b-5p causes a highly aggressive myeloid leukemia related to targeting the gene Lin28A (53). MYC is a basic transcription factor of the helix-loop-helix-leucine zipper family, which is found upregulated in AML and has been shown to block the myeloid cell differentiation (33). Retroviral transduction of MYC into murine bone marrow cells results in AML development (33,54). Similarly, sustained expression of a human MYC transgene culminated in the formation of AML in mice (33). NFKB1 is a transcription factor that is constitutively activated in primitive human AML cells, while inhibition of NFKB1 displayed a rapid induction of apoptosis in AML cells (32,55). After analyzing the topology of subnetwork, we found that TCF3 was a potential key regulator in our network due to its direct linking to all the four hubs in the subnetworks (Fig. 5), which may establish cross-talk among these subnetworks. In particular, TCF3 was predicted to regulate 17 target genes which have been involved in AML pathway in the four subnetworks. As a member of the TCF/LEF transcription factor family, TCF3 is essential for development, lymphopoiesis and stem cell function (44,56). Emerging evidence suggests that TCF3 is involved in the pathogenesis of several types of human cancer including colorectal cancer, prostate cancer and breast cancer (5759). Furthermore, the t(1;19) translocation between TCF3 and PBX1 genes causes expression of a TCF3-PBX1 fusion protein, and has been identified in B-cell acute lymphoblastic leukemia (B-ALL), which established a potential relationship between TCF3 and the etiology of B-ALL (44). However, the exact molecular mechanism of TCF3 as it relates to AML occurrence and development is still unknown.

In the subnetworks, TCF3 was potentially targeted by miR-15a-5p, MYC and NFKB1, and also targeting miR-125b-5p, MYC, ERG and PIM1 (Fig. 5). ERG is a megakaryocytic oncogene, which is required for normal megakaryopoiesis and play crucial role in establishing definitive hematopoiesis (60). A high expression level of ERG in AML is associated with poor prognosis (61). Transgenic expression of the human ERG causes early progenitor myeloid leukemia in mice similar to human AML, which was mediated by inducing the expression of the oncogenic kinase PIM1 through binding to a novel 3′ enhancer (62). PIM1 is a constitutively active serine/threonine protein kinase that normally functions in the proliferation and survival of hematopoietic cells in response to cytokines and growth factors (63). Overexpression of PIM1 has been detected in a range of solid cancers and hematologic malignancies (64). In AML, PIM1 has been suggested to have important roles in cell proliferation, survival, homing and migration, and the prognostic relevance of PIM1 upregulation was also observed in high ERG-expressing AML by the significantly decreased survival of these patients (62). In addition, ERG was the predicted target of MYC, NFKB1 and TCF3, while PIM1 was predicted to be regulated by miR-15a-5p, MYC and TCF3 in our subnetworks (Fig. 5). The above discussion led us to present a model of the involvement of TCF3 and the four hub nodes in subnetworks within AML (Fig. 6). According to the literature surveys and our subnetwork analysis, we propose that TCF3 is a potential key regulator in the miRNA-TF regulatory network linked to AML, which may play its regulatory role via miR-15a-5p, miR-125b-5p, MYC, NFKB1, ERG or PIM1.

In summary, this is the first study that describes the global expression profiling of miRNAs and TFs relating to AML using miRNA sequencing and TF array technology, respectively. Applying systems biology approaches to integrate the experimental data to multiple types of computational prediction on gene regulation, we constructed a miRNA-TF regulatory network specifically for AML, which may provide some hub regulators and clues for exploring the molecular mechanisms of AML. Furthermore, the present study also provides potential therapeutic targets for AML and proposes directions for further experimental research.

Acknowledgements

We sincerely thank all donors who participated in this investigation.

Abbreviations:

AML

acute myeloid leukemia

miRNA

microRNA

TF

transcription factor

pri-miRNA

primary miRNA

pre-miRNA

precursor miRNA

3′-UTR

3′-untranslated region

TFBS

transcription factor binding site

TSS

transcription start sites

GO

Gene Oncology

KEGG pathway

Kyoto Encydopedia of Gene and Genomes

qPCR

quantitative PCR

PAGE

polyacrylamide gel electrophoresis

HRP

streptavidin-conjugated horseradish peroxidase

lncRNA

long non-coding RNA

FBL

feedback loop

FFL

feed-forward loop

DAVID

the database for annotation, visualization and integrated discovery

References

1 

Conway O'Brien E, Prideaux S and Chevassut T: The epigenetic landscape of acute myeloid leukemia. Adv Hematol. 2014:1031752014.PubMed/NCBI

2 

Lee HJ, Daver N, Kantarjian HM, Verstovsek S and Ravandi F: The role of JAK pathway dysregulation in the pathogenesis and treatment of acute myeloid leukemia. Clin Cancer Res. 19:327–335. 2013. View Article : Google Scholar

3 

Zaidi SK, Trombly DJ, Dowdy CR, Lian JB, Stein JL, van Wijnen AJ and Stein GS: Epigenetic mechanisms in leukemia. Adv Biol Regul. 52:369–376. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Jamieson K and Odenike O: Late-phase investigational approaches for the treatment of relapsed/refractory acute myeloid leukemia. Expert Opin Pharmacother. 13:2171–2187. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Sabnis H, Bradley HL, Bunting ST, Cooper TM and Bunting KD: Capillary nano-immunoassay for Akt 1/2/3 and 4EBP1 phosphorylation in acute myeloid leukemia. J Transl Med. 12:1662014. View Article : Google Scholar : PubMed/NCBI

6 

Naoe T and Kiyoi H: Gene mutations of acute myeloid leukemia in the genome era. Int J Hematol. 97:165–174. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Larsson CA, Cote G and Quintás-Cardama A: The changing mutational landscape of acute myeloid leukemia and myelodysplastic syndrome. Mol Cancer Res. 11:815–827. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Abdel-Wahab O and Levine RL: Mutations in epigenetic modifiers in the pathogenesis and therapy of acute myeloid leukemia. Blood. 121:3563–3572. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Yan S, Yim LY, Lu L, Lau CS and Chan VS: MicroRNA regulation in systemic lupus erythematosus pathogenesis. Immune Netw. 14:138–148. 2014. View Article : Google Scholar : PubMed/NCBI

10 

van Rooij E and Kauppinen S: Development of microRNA therapeutics is coming of age. EMBO Mol Med. 6:851–864. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Babashah S, Sadeghizadeh M, Tavirani MR, Farivar S and Soleimani M: Aberrant microRNA expression and its implications in the pathogenesis of leukemias. Cell Oncol (Dordr). 35:317–334. 2012. View Article : Google Scholar

12 

Marcucci G, Mrózek K, Radmacher MD, Garzon R and Bloomfield CD: The prognostic and functional role of microRNAs in acute myeloid leukemia. Blood. 117:1121–1129. 2011. View Article : Google Scholar :

13 

Han YC, Park CY, Bhagat G, Zhang J, Wang Y, Fan JB, Liu M, Zou Y, Weissman IL and Gu H: microRNA-29a induces aberrant self-renewal capacity in hematopoietic progenitors, biased myeloid development, and acute myeloid leukemia. J Exp Med. 207:475–489. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Bai J, Guo A, Hong Z and Kuai W: Upregulation of microRNA-100 predicts poor prognosis in patients with pediatric acute myeloid leukemia. Onco Targets Ther. 5:213–219. 2012.PubMed/NCBI

15 

Wang Z, Hong Z, Gao F and Feng W: Upregulation of microRNA-375 is associated with poor prognosis in pediatric acute myeloid leukemia. Mol Cell Biochem. 383:59–65. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Li Y, Gao L, Luo X, Wang L, Gao X, Wang W, Sun J, Dou L, Li J, Xu C, et al: Epigenetic silencing of microRNA-193a contributes to leukemogenesis in t(8;21) acute myeloid leukemia by activating the PTEN/PI3K signal pathway. Blood. 121:499–509. 2013. View Article : Google Scholar

17 

Hickey CJ, Schwind S, Radomska HS, Dorrance AM, Santhanam R, Mishra A, Wu YZ, Alachkar H, Maharry K, Nicolet D, et al: Lenalidomide-mediated enhanced translation of C/EBPα-p30 protein up-regulates expression of the antileukemic microRNA-181a in acute myeloid leukemia. Blood. 121:159–169. 2013. View Article : Google Scholar :

18 

Zhang HM, Kuang S, Xiong X, Gao T, Liu C and Guo AY: Transcription factor and microRNA co-regulatory loops: Important regulatory motifs in biological processes and diseases. Brief Bioinform. 16:45–58. 2015. View Article : Google Scholar

19 

Cheng C, Yan KK, Hwang W, Qian J, Bhardwaj N, Rozowsky J, Lu ZJ, Niu W, Alves P, Kato M, et al: Construction and analysis of an integrated regulatory network derived from high-throughput sequencing data. PLoS Comput Biol. 7:e10021902011. View Article : Google Scholar : PubMed/NCBI

20 

Pulikkan JA, Dengler V, Peramangalam PS, Peer Zada AA, Müller-Tidow C, Bohlander SK, Tenen DG and Behre G: Cell-cycle regulator E2F1 and microRNA-223 comprise an auto-regulatory negative feedback loop in acute myeloid leukemia. Blood. 115:1768–1778. 2010. View Article : Google Scholar :

21 

Katzerke C, Madan V, Gerloff D, Bräuer-Hartmann D, Hartmann JU, Wurm AA, Müller-Tidow C, Schnittger S, Tenen DG, Niederwieser D, et al: Transcription factor C/EBPα-induced microRNA-30c inactivates Notch1 during granulopoiesis and is downregulated in acute myeloid leukemia. Blood. 122:2433–2442. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Padró T, Ruiz S, Bieker R, Bürger H, Steins M, Kienast J, Büchner T, Berdel WE and Mesters RM: Increased angiogenesis in the bone marrow of patients with acute myeloid leukemia. Blood. 95:2637–2644. 2000.

23 

Aurelius J, Martner A, Brune M, Palmqvist L, Hansson M, Hellstrand K and Thoren FB: Remission maintenance in acute myeloid leukemia: Impact of functional histamine H2 receptors expressed by leukemic cells. Haematologica. 97:1904–1908. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Pfaffl MW: A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res. 29:e452001. View Article : Google Scholar : PubMed/NCBI

25 

Sui W, Lin H, Peng W, Huang Y, Chen J, Zhang Y and Dai Y: Molecular dysfunctions in acute rejection after renal transplantation revealed by integrated analysis of transcription factor, microRNA and long noncoding RNA. Genomics. 102:310–322. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Lin Y, Wu J, Chen H, Mao Y, Liu Y, Mao Q, Yang K, Zheng X and Xie L: Cyclin-dependent kinase 4 is a novel target in micoRNA-195-mediated cell cycle arrest in bladder cancer cells. FEBS Lett. 586:442–447. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Rappaport N, Twik M, Nativ N, Stelzer G, Bahir I, Stein TI, Safran M and Lancet D: MalaCards: A comprehensive automatically-mined database of human diseases. Curr Protoc Bioinformatics. 47:1.24.1–1.24.19. 2014. View Article : Google Scholar

28 

Felice B, Cattoglio C, Cittaro D, Testa A, Miccio A, Ferrari G, Luzi L, Recchia A and Mavilio F: Transcription factor binding sites are genetic determinants of retroviral integration in the human genome. PLoS One. 4:e45712009. View Article : Google Scholar : PubMed/NCBI

29 

Huang DW, Sherman BT, Tan Q, Kir J, Liu D, Bryant D, Guo Y, Stephens R, Baseler MW, Lane HC, et al: DAVID Bioinformatics Resources: Expanded annotation database and novel algorithms to better extract biology from large gene lists. Nucleic Acids Res. 35(Web Server): W169–W175. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Li Z, Lu J, Sun M, Mi S, Zhang H, Luo RT, Chen P, Wang Y, Yan M, Qian Z, et al: Distinct microRNA expression profiles in acute myeloid leukemia with common translocations. Proc Natl Acad Sci USA. 105:15535–15540. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Fischer J, Rossetti S, Datta A, Eng K, Beghini A and Sacchi N: miR-17 deregulates a core RUNX1-miRNA mechanism of CBF acute myeloid leukemia. Mol Cancer. 14:72015. View Article : Google Scholar : PubMed/NCBI

32 

Grosjean-Raillard J, Adès L, Boehrer S, Tailler M, Fabre C, Braun T, De Botton S, Israel A, Fenaux P and Kroemer G: Flt3 receptor inhibition reduces constitutive NFkappaB activation in high-risk myelodysplastic syndrome and acute myeloid leukemia. Apoptosis. 13:1148–1161. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Brondfield S, Umesh S, Corella A, Zuber J, Rappaport AR, Gaillard C, Lowe SW, Goga A and Kogan SC: Direct and indirect targeting of MYC to treat acute myeloid leukemia. Cancer Chemother Pharmacol. 76:35–46. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Mott JL, Kurita S, Cazanave SC, Bronk SF, Werneburg NW and Fernandez-Zapico ME: Transcriptional suppression of mir-29b-1/mir-29a promoter by c-Myc, hedgehog, and NF-kappaB. J Cell Biochem. 110:1155–1164. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Tao J, Zhao X and Tao J: c-MYC-miRNA circuitry: A central regulator of aggressive B-cell malignancies. Cell Cycle. 13:191–198. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Steffen B, Müller-Tidow C, Schwäble J, Berdel WE and Serve H: The molecular pathogenesis of acute myeloid leukemia. Crit Rev Oncol Hematol. 56:195–221. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Schimmer AD: Novel therapies targeting the apoptosis pathway for the treatment of acute myeloid leukemia. Curr Treat Options Oncol. 8:277–286. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Kim HS, Lim GY, Hwang J, Ryoo ZY, Huh TL and Lee S: Induction of apoptosis by obovatol as a novel therapeutic strategy for acute myeloid leukemia. Int J Mol Med. 34:1675–1680. 2014.PubMed/NCBI

39 

Mikesch JH, Steffen B, Berdel WE, Serve H and Müller-Tidow C: The emerging role of Wnt signaling in the pathogenesis of acute myeloid leukemia. Leukemia. 21:1638–1647. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Li L, Tan Y, Chen X, Xu Z, Yang S, Ren F, Guo H, Wang X, Chen Y, Li G, et al: MDM4 overexpressed in acute myeloid leukemia patients with complex karyotype and wild-type TP53. PLoS One. 9:e1130882014. View Article : Google Scholar : PubMed/NCBI

41 

Chim CS, Wong AS and Kwong YL: Epigenetic inactivation of INK4/CDK/RB cell cycle pathway in acute leukemias. Ann Hematol. 82:738–742. 2003. View Article : Google Scholar : PubMed/NCBI

42 

Fukuda Y, Lian S and Schuetz JD: Leukemia and ABC transporters. Adv Cancer Res. 125:171–196. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Li YJ, Higgins RR, Pak BJ, Shivdasani RA, Ney PA, Archer M and Ben-David Y: p45NFE2 is a negative regulator of erythroid proliferation which contributes to the progression of friend virus-induced erythroleukemias. Mol Cell Biol. 21:73–80. 2001. View Article : Google Scholar

44 

Somasundaram R, Prasad MA, Ungerbäck J and Sigvardsson M: Transcription factor networks in B-cell differentiation link development to acute lymphoid leukemia. Blood. 126:144–152. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Zada AA, Pulikkan JA, Bararia D, Geletu M, Trivedi AK, Balkhi MY, Hiddemann WD, Tenen DG, Behre HM and Behre G: Proteomic discovery of Max as a novel interacting partner of C/EBPalpha: A Myc/Max/Mad link. Leukemia. 20:2137–2146. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Pellicano F, Scott MT, Helgason GV, Hopcroft LE, Allan EK, Aspinall-O'Dea M, Copland M, Pierce A, Huntly BJ, Whetton AD, et al: The antiproliferative activity of kinase inhibitors in chronic myeloid leukemia cells is mediated by FOXO transcription factors. Stem Cells. 32:2324–2337. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Gao SM, Xing CY, Chen CQ, Lin SS, Dong PH and Yu FJ: miR-15a and miR-16-1 inhibit the proliferation of leukemic cells by down-regulating WT1 protein level. J Exp Clin Cancer Res. 30:1102011. View Article : Google Scholar : PubMed/NCBI

48 

Cho JH, Gelinas R, Wang K, Etheridge A, Piper MG, Batte K, Dakhallah D, Price J, Bornman D, Zhang S, et al: Systems biology of interstitial lung diseases: Integration of mRNA and microRNA expression changes. BMC Med Genomics. 4:82011. View Article : Google Scholar : PubMed/NCBI

49 

Ye H, Liu X, Lv M, Wu Y, Kuang S, Gong J, Yuan P, Zhong Z, Li Q, Jia H, et al: MicroRNA and transcription factor co-regulatory network analysis reveals miR-19 inhibits CYLD in T-cell acute lymphoblastic leukemia. Nucleic Acids Res. 40:5201–5214. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Yan Z, Shah PK, Amin SB, Samur MK, Huang N, Wang X, Misra V, Ji H, Gabuzda D and Li C: Integrative analysis of gene and miRNA expression profiles with transcription factor-miRNA feed-forward loops identifies regulators in human cancers. Nucleic Acids Res. 40:e1352012. View Article : Google Scholar : PubMed/NCBI

51 

Gao SM, Xing CY, Chen CQ, Lin SS, Dong PH and Yu FJ: miR-15a and miR-16-1 inhibit the proliferation of leukemic cells by down-regulating WT1 protein level. J Exp Clin Cancer Res. 30:1102011. View Article : Google Scholar : PubMed/NCBI

52 

Aqeilan RI, Calin GA and Croce CM: miR-15a and miR-16-1 in cancer: Discovery, function and future perspectives. Cell Death Differ. 17:215–220. 2010. View Article : Google Scholar

53 

Chaudhuri AA, So AY, Mehta A, Minisandram A, Sinha N, Jonsson VD, Rao DS, O'Connell RM and Baltimore D: Oncomir miR-125b regulates hematopoiesis by targeting the gene Lin28A. Proc Natl Acad Sci USA. 109:4233–4238. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Luo H, Li Q, O'Neal J, Kreisel F, Le Beau MM and Tomasson MH: c-Myc rapidly induces acute myeloid leukemia in mice without evidence of lymphoma-associated antiapoptotic mutations. Blood. 106:2452–2461. 2005. View Article : Google Scholar : PubMed/NCBI

55 

Guzman ML, Neering SJ, Upchurch D, Grimes B, Howard DS, Rizzieri DA, Luger SM and Jordan CT: Nuclear factor-kappaB is constitutively activated in primitive human acute myelogenous leukemia cells. Blood. 98:2301–2307. 2001. View Article : Google Scholar : PubMed/NCBI

56 

Howard JM, Nuguid JM, Ngole D and Nguyen H: Tcf3 expression marks both stem and progenitor cells in multiple epithelia. Development. 141:3143–3152. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Slyper M, Shahar A, Bar-Ziv A, Granit RZ, Hamburger T, Maly B, Peretz T and Ben-Porath I: Control of breast cancer growth and initiation by the stem cell-associated transcription factor TCF3. Cancer Res. 72:5613–5624. 2012. View Article : Google Scholar : PubMed/NCBI

58 

Li C, Cai S, Wang X and Jiang Z: Hypomethylation-associated up-regulation of TCF3 expression and recurrence in stage II and III colorectal cancer. PLoS One. 9:e1120052014. View Article : Google Scholar : PubMed/NCBI

59 

Patel D and Chaudhary J: Increased expression of bHLH transcription factor E2A (TCF3) in prostate cancer promotes proliferation and confers resistance to doxorubicin induced apoptosis. Biochem Biophys Res Commun. 422:146–151. 2012. View Article : Google Scholar : PubMed/NCBI

60 

Salek-Ardakani S, Smooha G, de Boer J, Sebire NJ, Morrow M, Rainis L, Lee S, Williams O, Izraeli S and Brady HJ: ERG is a megakaryocytic oncogene. Cancer Res. 69:4665–4673. 2009. View Article : Google Scholar : PubMed/NCBI

61 

Marcucci G, Maharry K, Whitman SP, Vukosavljevic T, Paschka P, Langer C, Mrózek K, Baldus CD, Carroll AJ, Powell BL, et al; Cancer and Leukemia Group B Study. 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. 25:3337–3343. 2007. View Article : Google Scholar : PubMed/NCBI

62 

Goldberg L, Tijssen MR, Birger Y, Hannah RL, Kinston SJ, Schütte J, Beck D, Knezevic K, Schiby G, Jacob-Hirsch J, et al: Genome-scale expression and transcription factor binding profiles reveal therapeutic targets in transgenic ERG myeloid leukemia. Blood. 122:2694–2703. 2013. View Article : Google Scholar : PubMed/NCBI

63 

Burger MT, Han W, Lan J, Nishiguchi G, Bellamacina C, Lindval M, Atallah G, Ding Y, Mathur M, McBride C, et al: Structure guided optimization, in vitro activity, and in vivo activity of Pan-PIM kinase inhibitors. ACS Med Chem Lett. 4:1193–1197. 2013. View Article : Google Scholar

64 

Shah N, Pang B, Yeoh KG, Thorn S, Chen CS, Lilly MB and Salto-Tellez M: Potential roles for the PIM1 kinase in human cancer - a molecular and therapeutic appraisal. Eur J Cancer. 44:2144–2151. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2016
Volume 48 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lin X, Xu Y, Sun G, Wen J, Li N, Zhang Y, Yang Z, Zhang H and Dai Y: Molecular dysfunctions in acute myeloid leukemia revealed by integrated analysis of microRNA and transcription factor. Int J Oncol 48: 2367-2380, 2016
APA
Lin, X., Xu, Y., Sun, G., Wen, J., Li, N., Zhang, Y. ... Dai, Y. (2016). Molecular dysfunctions in acute myeloid leukemia revealed by integrated analysis of microRNA and transcription factor. International Journal of Oncology, 48, 2367-2380. https://doi.org/10.3892/ijo.2016.3489
MLA
Lin, X., Xu, Y., Sun, G., Wen, J., Li, N., Zhang, Y., Yang, Z., Zhang, H., Dai, Y."Molecular dysfunctions in acute myeloid leukemia revealed by integrated analysis of microRNA and transcription factor". International Journal of Oncology 48.6 (2016): 2367-2380.
Chicago
Lin, X., Xu, Y., Sun, G., Wen, J., Li, N., Zhang, Y., Yang, Z., Zhang, H., Dai, Y."Molecular dysfunctions in acute myeloid leukemia revealed by integrated analysis of microRNA and transcription factor". International Journal of Oncology 48, no. 6 (2016): 2367-2380. https://doi.org/10.3892/ijo.2016.3489