Buscar en
Boletín Médico del Hospital Infantil de México (English Edition)
Toda la web
Inicio Boletín Médico del Hospital Infantil de México (English Edition) Epigenetic alterations in acute lymphoblastic leukemia
Journal Information
Vol. 74. Issue 4.
Pages 243-264 (July - August 2017)
Visits
10581
Vol. 74. Issue 4.
Pages 243-264 (July - August 2017)
Review article
Open Access
Epigenetic alterations in acute lymphoblastic leukemia
Alteraciones epigenéticas en leucemia linfoblástica aguda
Visits
10581
María del Pilar Navarrete-Meneses, Patricia Pérez-Vera
Corresponding author
pperezvera@yahoo.com

Corresponding author.
Laboratory of Genetics and Cancer, Department of Human Genetics, Instituto Nacional de Pediatría, Mexico City, Mexico
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (4)
Show moreShow less
Tables (6)
Table 1. Hypermethylated genes in acute lymphoblastic leukemia, identified through directed methylation studies.
Table 2. Altered methylation/expression profiles in acute lymphoblastic leukemia identified through genome wide association studies.
Table 3. Methylation profile of the ETV6-RUNX1 subtype in pediatric ALL-B.
Table 4. Methylation profile of the hyperdiploid subtype in pediatric ALL-B.
Table 5. Methylation profile of other subtypes of pediatric ALL-B.
Table 6. Target genes of frequently altered miRNAs in ALL.
Show moreShow less
Abstract

Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. It is well-known that genetic alterations constitute the basis for the etiology of ALL. However, genetic abnormalities are not enough for the complete development of the disease, and additional alterations such as epigenetic modifications are required. Such alterations, like DNA methylation, histone modifications, and noncoding RNA regulation have been identified in ALL. DNA hypermethylation in promoter regions is one of the most frequent epigenetic modifications observed in ALL. This modification frequently leads to gene silencing in tumor suppressor genes, and in consequence, contributes to leukemogenesis. Alterations in histone remodeling proteins have also been detected in ALL, such as the overexpression of histone deacetylases enzymes, and alteration of acetyltransferases and methyltransferases. ALL also shows alteration in the expression of miRNAs, and in consequence, the modification in the expression of their target genes. All of these epigenetic modifications are key events in the malignant transformation since they lead to the deregulation of oncogenes as BLK, WNT5B and WISP1, and tumor suppressors such as FHIT, CDKN2A, CDKN2B, and TP53, which alter fundamental cellular processes and potentially lead to the development of ALL. Both genetic and epigenetic alterations contribute to the development and evolution of ALL.

Keywords:
Acute lymphoblastic leukemia
Epigenetics
Gene expression
DNA methylation
Histone modifications
miRNAs
Resumen

La leucemia linfoblástica aguda (LLA) es el tipo de cáncer más frecuente en niños. Aunque se sabe que las alteraciones genéticas constituyen la base de la etiología de la LLA, se ha demostrado que no son suficientes para el desarrollo leucémico; son necesarias alteraciones adicionales, como las modificaciones epigenéticas. En la LLA se han identificado alteraciones de este tipo, como la metilación del DNA, la modificación de histonas y la regulación por RNAs no codificantes. La hipermetilación del DNA en regiones promotoras es una de las alteraciones epigenéticas más frecuentes en LLA: conlleva al silenciamiento de genes que generalmente son supresores de tumor y, en consecuencia, contribuye a la leucemogénesis. También se han detectado alteraciones en proteínas remodeladoras de histonas, como la sobreexpresión de enzimas desacetilasas de histonas, así como alteraciones en enzimas acetil transferasas y metil transferasas. En la LLA también se altera la expresión de miRNAs, lo cual produce desregulación en la expresión de sus genes blanco. Estas modificaciones epigenéticas son eventos clave en la transformación maligna, e involucran la desregulación de oncogenes como BLK, WNT5B y WISP1 y de supresores de tumor como FHIT, CDKN2A, CDKN2B y TP73, lo que afecta diversos procesos celulares fundamentales que conllevan al desarrollo de LLA. Las alteraciones epigenéticas y genéticas contribuyen en conjunto al desarrollo y evolución de la LLA.

Palabras clave:
Leucemia linfoblástica aguda
Epigenética
Expresión génica
Metilación del DNA
Modificaciones de histonas
miRNAs
Full Text
1Introduction

Leukemias are the most common pediatric neoplasms; acute lymphoblastic leukemia (ALL) is the most frequent type.1 Diverse specific genetic alterations have been identified in ALL that lead to the development of this disease.1,2 However, it is well recognized that these are not sufficient for leukemic transformation. Therefore, additional events are required.3 Currently, it is known that—in addition to genetic alterations—epigenetic changes are also fundamental in the leukemogenic process.4–6

Epigenetics studies heritable changes that lead to modifications in gene expression, which not imply alterations to the DNA sequence.7,8 DNA methylation, histone modification, nucleosome remodeling and regulation of noncoding RNA are among the most studied epigenetic mechanisms (Figure 1).9,10 These epigenetic alterations constitute fundamental events in the development of ALL. In this review, the main epigenetic alterations found in ALL are presented and defined. DNA methylation, histone modification like acetylation, microRNA regulation, and genes modified by these mechanisms are included. Although epigenetic alterations are fundamental in ALL of B (ALL-B) and T (ALL-T) lineage in both adults and children, this review is focused mainly on pediatric ALL-B due to the high incidence of this disease in this age group. The study of epigenetic modifications in ALL is of vital importance for knowledge concerning the etiology of the disease, as well as for the search of potential therapeutic targets.4,9

Figure 1.

Levels of epigenetic regulation. Epigenetic regulation involves mechanisms such as DNA methylation (a), histone modification (b), and regulation by non-coding RNAs (c). In each case, the conditions under gene expression is induced (green) or silenced (red) are indicated.

(0.44MB).
2Acute lymphoblastic leukemia

ALL is characterized by the uncontrolled proliferation of immature lymphoid cells called lymphoblasts, which predominate in the bone marrow and alter normal hematopoiesis.2,11 ALL affects T or B lineage lymphocytes and occurs in adults and children.11 It is of special relevance in the pediatric stage since it is the most common cancer, representing 25% of the cases. Eighty-five percent of ALL cases in children are of the B precursor type (ALL-B). Pediatric ALL cases are phenotypically and genetically heterogeneous and the etiology has not been completely determined.12

ALL has been classified in subgroups according to the presence of genetic alterations, which have a strong influence on the patient's prognosis.13 Approximately 75% of pediatric ALL cases present some type of numerical or structural chromosomic alteration.14 The most common numeric alteration in pediatric ALL is hyperdiploidy15; translocations are the most frequent chromosomal structural alterations16. The t(12;21)(p13;q22) translocation generates the fusion of ETV6-RUNX1 genes and is present in 20-25% of the cases.15,16 The t(1;19)(q23;p13) translocation leads to the fusion of TCF3-PBX1 genes and is present in 5-6% of the cases of pediatric ALL.13 The t(9;22)(q34;q11) translocation generates the fusion of BCR-ABL1 genes and is present in 3% of the cases.11,16 Translocations involving the MLL gene (KMT2A) are detected in 3% of patients between 2-5 years of age and 80% of those under one year of age. Although these genetic alterations are capable of starting the process of leukemogenesis, they are generally not sufficient; hence, additional genetic or epigenetic modifications are necessary for the development and evolution of ALL.16

3DNA methylation

DNA methylation is the most studied epigenetic mechanism in ALL.4 This modification involves the covalent addition of methyl groups to the carbon 5 of the DNA's cytosine. In mammals, DNA methylation occurs primarily in the sequence 5′-CpG-3′, known as CpG island.17 CpG dinucleotides are found in low density throughout the genome; however, there is a high density of this sequence in the promoter regions.17 Approximately, 70% of the promoter regions in mammals present CpG islands.10 DNA methylation of the promoting regions generally correlates with gene expression silencing.17 On the contrary, CpG methylation along the body of the gene promotes elongation during transcription.8

The enzymes responsible of adding the methyl groups to the DNA are known as DNA methyltransferases (DNMTs), and at least three have been discovered in eukaryotes.18 The DNMT1 is in charge of maintaining DNA methylation during replication: it methylates newly synthesized CpG dinucleotides.10,18 The DNMT3a and DNMT3b establish, primarily, de novo methylation during embryogenesis8,10; additionally, they repair errors by the DNMT1 during DNA synthesis.8 In contrast, demethylation of the DNA involves the enzyme TET2 (ten-eleven-translocation 2), which catalyzes the conversion of 5-methylcytosine to 5-hidroxymethylcytosine.19

DNA methylation is one of the most studied epigenetic modifications in cancer. In some types of cancer, tumor cells show a global hypomethylation of the DNA, which is associated with chromosomal instability, transposon reactivation, and loss of the genomic imprint.4,8 In general, cancer cells present hypermethylation of the DNA in promoter regions. It has been reported that abnormal methylation in cancer is present in 5-10% of the normally demethylated promoters.10 This abnormal hypermethylation can lead to silencing of tumor suppressor genes, and consequently to a neoplastic process.18 Abnormal hypermethylation not only affects coding gene expression but also noncoding RNA expression, which can contribute to malignant transformation.10 Alteration of DNA methylation is a highly prevalent event in several types of cancer, including ALL (Figure 2).

Figure 2.

Epigenetic differences between normal lymphoid progenitor cells and leukemic cells.

(0.13MB).
3.1Alterations in DNA methylation in ALL3.1.1Directed studies

Numerous studies have established that disruption of DNA methylation has a fundamental role in the development of ALL in both B and T lineages in adults and children. The study of the alterations in gene methylation in ALL has been approached in two ways: through the directed study of specific genes preselected for their importance in ALL and through genome wide studies in which the analysis is not limited to certain genes.20 In most cases, the state of gene methylation has been tied with studies of gene expression. Altogether, these studies have revealed aberrant methylation and expression, mainly simultaneous hypermethylation in multiple genes, which has been defined as a “methylator phenotype”. This alteration leads to gene silencing and is a common characteristic of ALL and other types of leukemia in adults and children.4,20,21

Through directed studies, several hypermethylated genes have been found in patients with ALL (Table 1). Some of these include genes such as TP73, FHIT, MME (CD10), TET2, sFRP2, EFNA5, ESR1, THBS2, CDKN1A (p21), ABCB1, EPHB4, CDKN2B (p15), CDKN2A (p16), SYK, DAPK1, BRINP1 (DBC1), ABL1, PTEN, sFRP5, BNIP3, MYOD1, CALCA (calcitonin), CDKN1C (p57), DKK3, WIF1, LATS2, EFNB2, PPP1R13B (ASPP1), THBS1, CDH1, TP53 and HCK.22–46 Most of these tumor suppressor genes are involved in cellular functions and fundamental signaling pathways, such as those regulating the cell cycle, apoptosis, DNA-damage response, and gene expression (Table 1). Based on the results of these directed studies, several correlations have been observed between the patterns of methylation and the subtypes of ALL have been obtained, as well as the prognosis of the disease.

Table 1.

Hypermethylated genes in acute lymphoblastic leukemia, identified through directed methylation studies.

Gene  Chromosome  Cell function/implicated pathway  Type of patient  Lineage  Reference 
TP73  p53  P, A  B, T  García-Manero et al., 200329
Kawano et al., 199931
Corn et al.,199932
Shen et al., 200238
Gutiérrez et al.,200340
Canalli et al., 200541 
FHIT  Purine metabolism  B, T  Zheng et al., 200430 
MME (CD10)  NF-kappa B  P, A  B, T  García-Manero et al., 200329 
TET2  DNA demethylation    Bahari et al., 201639 
sFRP2  WNT pathway  B
ETV6-RUNX1 
San José-Enériz et al., 200645 
EPH/EPHRIN
EFNA5 family 
Family of epinephrine receptors and ligands  B, T  Kuang et al.,201036 
ESR1 (ER)  Estrogen receptor and gene transcription  P, A    Issa et al.,199635
García-Manero et al., 200224
Shen et al., 200238 
THBS2  Cell-cell interactions  B, T  García-Manero et al., 200224 
CDNK1A (p21)  Cell cycle  P, A  B, T  Román-Gómez et al., 200237* 
ABCB1 (MDR1)  ABC transporters  P, A  B, T  García-Manero et al.,200329 
EPH/EPHRIN
EPHB4 family 
Family of epinephrine receptors and ligands  B, T  Kuang et al., 201036 
CDKN2B (p15)  CDK kinases and cell cycle  P, A  B, T  Batova et al., 199722
Wong et al., 200023
García-Manero et al., 200224
Mai et al., 201625
Shen et al., 200238
Gutiérrez et al., 200340 
CDKN2A (p16)  CDK kinases and cell cycle  P, A  B, T  Batova et al., 199722
García-Manero et al., 200224
Mirebeau et al., 200626
Nakamura et al., 199934 
SYK  JAK/STAT  P, A  Jiménez-Velasco et al., 200543 
DAPK1  Apoptosis and autophagy  B, T  Gutiérrez et al., 200340 
BRINP1 (DBC1)  Cell proliferation and apoptosis  P, A  B, T  San José-Enériz et al., 200645 
ABL1  Cell division, adhesion, differentiation, responses to stress  B, T  García-Manero et al., 200224 
PTEN  10  Tumor suppressor and AKT/PKB pathway  B
ETV6-RUNX1 
San José-Enériz et al., 200645 
sFRP5  10  WNT pathway  P, A  Jiménez-Velasco et al., 200543 
BNIP3  10  Apoptosis  —  —  Murai et al., 200546 
MYOD1  11  Transcription factor  B,T  García-Manero et al., 200224 
CALCA (calcitonin)  11  Calcium regulation  P, A  T,B  Roman et al., 200128 
CDKN1C (p57)  11  Cell cycle    Gutiérrez et al., 200542 
DKK3  11  WNT pathway  B
ETV6-RUNX1 
San José-Enériz et al., 200645 
WIF1  12  WNT pathway  P, A  Jiménez-Velasco et al., 200543 
LATS2  13  Tumor suppressor  P, A  B,T  Jiménez-Velasco et al., 200543 
Familia EPH/EPHRIN
EFNB2 
13  Epinephrine family and tyrosine-kinase receptors  B,T  Kuang et al., 201036 
PPP1R13B (ASPP-1)  14  Apoptosis/p53  P, A  Jiménez-Velasco et al., 200543 
THBS1  15  Cell-cell interaction  B, T  García-Manero et al., 200224 
CDH1  16  Cell adhesion  B, T  García-Manero et al., 200329
Corn et al., 200033 
TP53  17  Tumor suppressor, apoptosis and cell cycle  —  B, T  Agirre et al., 200327 
HCK  20  Tyrosine-kinase Src family  —  B
BCR-ABL1 
Hoshino et al., 200744 
*

Additional studies did not find aberrant methylation in CDKN1A [Ref. 38].

P, pediatric; A, adult; B, B cells; T, T cells.

3.1.2Genome wide association studies

Genome wide association studies include an extensive analysis of the genome, as well as a greater number of patients.17,20 It is noteworthy that an association between directed studies and great scale analysis has not been found in all cases, possibly due to significant variations in the methodology and the size of the studied population.20

Genome wide association studies have compared samples of patients with ALL-B and leukemic cell lines with non-leukemic tissues and identified numerous changes in DNA methylation, mainly hypermethylation, in all subtypes of ALL-B.47,48 Some studies have focused in pediatric ALL or adult ALL and others have analyzed both groups. In most of these studies, patients with ALL-B and ALL-T have been included. Taylor et al. studied a limited number of patients, six children and four adults with ALL-B or T, and identified 11 genes (DCC, DLC1, DDX51, KCNK2, LRP1B, NKX6-1, NOPE, PCDHGA12, RPIB9, ABCB1, SLCA1) with aberrant methylation (Table 2) in comparison with normal cells of peripheral blood and bone marrow origin. In this study, no differences in methylation between ALL of B and T lineage were found, except in gene DDX51, which did not show methylation in ALL-T. Moreover, no differences between adults and children were found.49

Table 2.

Altered methylation/expression profiles in acute lymphoblastic leukemia identified through genome wide association studies.

Study  Studied population  Lineage  Altered methylation genes 
Taylor et al., 200749  Adults: 4
Children: 6 
B and T  11 hypermethylated genes:
DCC, DLC1, DDX51, KCNK2, LRP1B, NKX6-1, NOPE, PCDHGA12, RPIB9, ABCB1, SLCA1 
Kuang et al., 200850  Myeloid and lymphoid cell lines
Adults: 61 
B and T  15 hypermethylated genes:
GIPC2, RSPO1, MAGI1, CAST1, ADCY5, HSPA4L, OCLN, EFNA5, MSX2, GFPT2, GNA14, SALL1, MYO5B, ZNF382, MN1 
Figueroa et al., 201352  Children: 167  B (137) and T (30)  85 genes with alterations in methylation, common among all subtypes (73 hypermethylated, 12 hypomethylated*):
TIE1, LPPR4, ANP32E, *DQX1, *AUP1, *HTRA2, *KCNJ3, GRM7, CEP97, DNAJC19, SENP2, DGKG, CETN3, CAMLG, PCDHA11, PCDHB17, PCDHB11, PCDHB11, PCDHGA5, PCDHGB3, *GNPDA1, PROP1, CMAH, *HIST1H2BJ, *HIST1H2AG, *HIST1H4I, SMAP1, FUT9, ATG5, SLC22A2, HOXA5, HOXA6, CHCHD2, BPGM, AGPAT6, MOS, SDR16C5, WDR67, WDYHV1, ELAVL2, C9orf25, DNAI1, MCART1, TAF3, KIAA1279, H2AFY2, NDUFB8, C10orf26, GPR26, CCDC34, *ELF5, LRRC4C, ZBTB16, SIAE, SPA17, TMTC1, C12orf44, SKA3, MRP63, DHRS12, OXA1L, PABPN1, SLC8A3, SCG5, TGFB1I1, CNOT1, SPNS2, ACACA, TADA2A, LSM12, G6PC3, *GPRC5C, OSBPL1A, INO80C, ZNF560, ZNF582, DDRGK1, ITPA, PTPRT, *OK/SW-cl.69, MAP7D2, GSPT2, PABPC5, MCTS1, MGC16121 
Chatterton et al., 201451  Children: 69  370 genes with alteration in methylation (325 hypermethylated, 45 hypomethylated*) common among all subtypes:
ZFP112, ZFP42, ZNF285, ZNF300, ZNF385D, ZNF677, ZFP37, ZNF560, CXCL1, CXCL3, CXCL5, CXCL6, ADRA1A, ADRA2C, AGTR1, CASR, FPR2, FZD9, GALR1, GPR126, GPR26, GPR37, GPR6, GPR77, GRM1, GRM7, HTR4, LHCGR, OPRM1, P2RY2, *PTAFR, PTGFR, QRFPR, SSTR1, GRIA2, GRIA4, GRIK1, GRIK2, GRIK3, KCNB1, KCNB2, KCNG1, KCNH7, KCNK1, KCNK5, KCNN2, *KCNQ4, TRPC1, TRPC4, TRPM3, SLC19A3, SLC1A1, SLC22A3, SLC24A3, SLC34A2, SLC44A4, SLC5A7, SLC6A1, SLC6A15, SLC6A2, PCDHB15, PCDHB2, PCDHB3, PCDH20, CDH6, CDH8, IL1RN, IL7, *LTA, ACADL, BHMT, CASD1, CYP24A1, CYP7B1, DHCR24, DPYS, DPYSL4, ELOVL4, EPHX2, GALNT11, GCNT2, GDA, LPL, LRAT, ME1, NMNAT3, *NOX3, PDE10A, PDE1C, PDE4B, PDE4C, PLOD2, PNPLA3, PRTFDC1, PTGS2, RGS7, SMPD3, ST6GAL2, UGGT2, CHI3L2, EFEMP1, KL, LYZ, PDZRN3, PLA2G7, POGLUT1, SMPDL3A, *DNTT, RFX6, ABO, ADCY2, *ENPP3, ENTPD3, GNA14, SH3GL2, MACROD2, HS6ST3, BMP7, FGF10, FGF12, GRP, KITLG, NELL1, PTN, NRG1, PEX5L, *VDAC3, CACNA1A, CACNA1D, CFTR, GABRA2, GLRB, GPM6A, HCN1, RYR2, NALCN, *BLK, *HSPB8, PRKD1, MYO3A, MAPK15, PAK7, *DDR1, EPHA5, ERBB4, FGFR2, KDR, NR2F2, PGR, THRB, *ARPP21, *EVL, *FGD2, *KISS1, *S100A2, *SORBS3, *SPRR4, *DEFB129, *MBP, *SFTPB, *WISP1, *WNT5B, *INTS1, *TCL1A, *CLDN15, *GP9, *LY9, *RPH3AL, *XIRP1, *ASB10, *FAM49A, *KLHDC7A, *SH2D4B, *SRBD1, ACTA1, DES, HSPA2, MT2A, NEFM, NEFH, SLPI, VSNL1, LIN7A, TOM1L1, CPLX2, RAB32, ERC2, PDLIM3, STXBP6, GULP1, PARVA, PARVG, NOD2, PREX2, CTTNBP2, RHPN2, PRKCDBP, SPATA18, SPTSSB, ADCYAP1, CBLN4, COL14A1, CYR61, FAM19A4, FAM20A, FSTL5, IGFBP3, LAMA1, LAMA2, MAMDC2, MATN2, NPTX2, RLN1, RLN2, SASH1, SCG3, SEMA3C, SERPINA1, SST, TAC1, TGFBI, TMEM45B, VSTM2A, ZPBP2, ACTL6B, CENPH, KIF6, MIPOL1, MLF1, NOL10, PRDM5, SNRPN, SYCP1, TDRD5, CADM1, CD8B, CDCP1, CLDN11, CNTNAP2, CTNND2, DSCAML1, EPB41L3, FLRT2, LSAMP, MARVELD3, NTM, PCDH8, PKP2, STEAP4, SYN2, THEM4, TPBG, ALS2CR11, C10orf107, C11orf70, C1orf115, C20orf197, C3orf14, C6orf118, C7orf63, CCDC37, CWH43, EFCAB1, FAM83A, FAM84A, FBXO39, FEZF2, FIBIN, LCA5, LPPR1, LRIG3, PHF21B, RBM47, SLC35F3, TMEM171, TMEM74, TSPYL5, TTC22, UNC80, WDR17, XKR6, CTSG, EPHX1, ADAM12, ADAMTS18, ADAMTS5, CPXM2, HP, PAPPA, PCSK1, PCSK2, PRSS12, TLL1, EYA4, PPP2R3A, CDC14B, PTPRO, PTPRZ1, LPPR4, ACTN2, BHLHE22, BNC1, DMRT1, DMRT2, DMRT3, EGR4, FOXA2, FOXG1, GATA4, GCM2, GSC2, HLF, HOXA2, HOXD1, HOXD11, ID4, IRF6, KLF1, *LIMD1, MSX2, MYF5, NACC2, NFE2, NFIB, NKX2-1, NPAS2, ONECUT2, OTX2, OVOL2, PAWR, *POU2AF1, POU4F3, PROX1, PRRX1, SCRT1, SIX1, SNCAIP, SOX1, SOX14, SOX17, SOX9, *TCF3, TLX3, TP73, WWC1, WWTR1, YAP1, ZIC1, EIF5A2, *ANXA9, CXADR, DCC, *ENG, FCGR2A, GPC6, *LRP8, *SCARF1, SFRP1, *HBZ, *SNX8, TF, UPF3A, SORCS3, ATP8A2, *LBP PDPN, SNAP25, SYT9, FAM164A, MOSC2 

B, B cells; T, T cells.

Kuang et al. analyzed 23 cell lines of lymphoid and myeloid origin (MOLT4, Jurkat, Peer, T-ALL1, CEM, J-TAG, B-JAB, RS4, ALL1, Raji, REH, Ramos, K562, BV173, HL60, NB4, THP1, U937, ML1, OCI, HEL, MOLM13, KBM5R) and identified altered methylation in 65 genes. In 15 of these genes (GIPC2, RSPO1, MAGI1, CAST1, ADCY5, HSPA4L, OCLN, EFNA5, MSX2, GFPT2, GNA14, SALL1, MYO5B, ZNF382 and MN1) methylation was confirmed by analyzing 61 samples of adults with ALL-B and T (Table 2). The ontological analysis of these genes revealed enrichment of pathways involved in proliferation, cell growth and differentiation, apoptosis, gene expression regulation, DNA replication and repair, signal transduction, transport, and metabolism.50

In the study of Chatterton et al., in 2014, levels of expression and methylation in 69 children with ALL-B were associated. Hypermethylation and decreased gene expression in 325 genes was identified, as well as hypomethylation and increased gene expression in 45 genes. These results were compared with 42 non-leukemic bone marrow samples in which these modifications were not found (Table 2).51 Ontological analysis of these 370 genes showed greater representation of molecular pathways associated with hematologic neoplasms, such as cell to cell signaling and interaction, death and cell survival, as well as cell development. In particular, hypermethylation was found in genes codifying proteins involved in signaling of the glutamate receptor, G-protein signaling, and adenosine-monophosphate (cAMP) mediated signaling; suppression of several proteins involved in the cation transport was also found. Hypermethylation of these genes and its subsequent suppression are indicative of a reduction in the potential of cell-microenvironment interaction, as well as the apoptotic potential. In this study, alterations in methylation of genes associated with the leukemic process were also identified, such as chemokines CXCL1, CXCL3, CXCL5 and CXCL6 and cytokines LTA and IL-7, which genes showed hypo and hypermethylation, respectively. Additionally, 11 dysregulated kinase genes were identified: BLK, DDR1 and HSPB8, which showed hypomethylation and overexpression, and EPHA5, ERBB4, FGFRDZ, KDR, MAPKI5, MYO3A, PAK7 and PRKD1, which were found to be hypermethylated and, consequently, silenced. Alterations in factors of the WNT signaling pathway were also found: hypomethylation and overexpression of WNT5B and WISP1, as well as hypermethylation and low expression of SFRP1, PP2A, and SOX. The signature of methylation/expression was common in all ALL subtypes.51 This study also analyzed expression signatures dependent on the different subtypes of ALL and a comparison with non-leukemic samples was conducted. The study identified 55, 51, and 13 genes with epigenetic dysregulation associated with subtypes ETV6-RUNX1, hyperdiploidy and “other alterations” (this group included patients with fusion of TCF3-PBX1, BCR-ABL and MLL translocations), respectively (Tables 3–5).51

Table 3.

Methylation profile of the ETV6-RUNX1 subtype in pediatric ALL-B.

Hypermethylated genes  Reference 
NRG1, KCNC3, RAB37, COL7A1, VIPR2, NIPAL1, GREM1, TM6SF1, MTX2, DPP4, DOCK5, SYT17, WWTR1, ITGAX, TMEM144, ASNS, MYO3A, GUCY1B2, CHD5, ITGB5, PAX7, CD44, RAB6B, PI4K2A, ADRA1D, FAM123A, STBD1, OVOL2, RNF207, CRABP1, CDC42BPB, L3MBTL4, KIAA0020, MPZL3, PRICKLE1, ADPRH, PDCD1LG2, SLCO2A1, FXYD6, VAX2, GSX2, SYT12, TMEM67, SLAMF8, CTTN, LPXN  Chatterton et al., 201451 
MYO1E, STBD1, OPHN1, HOXD1, WWTR1, SOX3, SNAI3, ZNF462, C4orf31, LRRC55, ST6GALNAC4, C21orf15, ALX1, GUCY2D, FOXA2, ZNF462, SIGLEC9, MTMR11, RIN1, PDE3A, UCN2, KCNN2, NKX2-8, PDK4, NHS, HOXD1, TNFRSF9, NEFL, C20orf165, NEURL2, CTSA, BMP7, SLC35B1, ITIH4, MUSTN1, SLC2A5, MEFV, PLD4, CXCR3, DARC, C9orf79, STBD1, PDK4, UCN2, SRGN, DBX2, C10orf116, AGAP11, ZNF215, BGLAP, PCDHGB4, PCDHGA8, BMP7, PJA2, GPR50, ESX1, KRT80, ZNF804A, TFCP2L1, GNPNAT1, OGG1, ADCY3, CBLN4, CHAT, SLC18A3, PLEKHG4, WNT2, C20orf165, NEURL2, CTSA, GPR123, SYT15, FCN1, BDNF, FBXW5, C8G, C14orf49, EFEMP1, BBC3, LEPREL2, PCDHGB2, PCDHGA5, PCDHGB3, CRYBB3, PTMS, LAG3, FAM190A, EIF2C2, PCDHGA5, PCDHGB3, NEFM, CCBL1, LRRC8A, TUB, TJP1, LIMD1, PPARD, MGC16703, P2RX6, FBXW5, C8G, ZNF296, GEMIN7, GRIA3, NOS3, GABRE, RTDR1, RAB36, ZNF19, MSX2, TSEN2, PRDM11, EPHA5, GPR81, S100A9, EIF6, RPL9, LIAS, SCARF2, CUEDC1, DOK5, HECW1, GPR126, PRKAG3, MARCKSL1, HIST3H2A, HIST3H2BB, SIAE, SPA17, MAGEB6, C14orf49, DAAM2, EIF2C1, TSPAN15, MSRB3, ITGAE, HLF  Figueroa et al., 201352 
GRHL3, CTTN  Nordlund et al., 201553 
CLIC5, ACVR1C, IGF2BP1, DSC2, POLO  Lee et al., 201555 
Hypomethylated genes   
SLC35B3, EPOR, BEST3, APOBEC3A, MPP7, TMED6, TRIM69, ALPK1, MPP7, CRYBB3  Chatterton et al., 201451 
MIB1, GFAP, TMED6, TPMT, KDM1B, GFAP, E2F6, DRD5, NCKAP1, CSTF3, HTR3C, HTR3C, FUCA1, DNAJC10, B4GALT3, BEST3, CALD1, SIDT1, LRRC4, GNB4, FCHSD2, ABHD3, RAD50, KCNK15, PIK3CG, LRRC4, C22orf24, YWHAH, RRAGA, NACA, VPS13C, SH2D3A, VAV1, C12orf49, RNFT2, ITSN2, TREML1, PRSS1, RAB3GAP1, MRPS15, ATF7, TNK1, ETV7, HLA-DQA1, ERLIN2, ST7L, CAPZA1, TUSC3, BAT2D1, LOC100127888, EID3, TSPYL1, DSE, ISG20, UFC1, ZNF143, SPEM1, C17orf74, C9orf40, MOBKL3, CAB39L, SETDB2, CSTF3, SLC35B3, ATP5J, GABPA, KCTD17, KLHDC5, TOR1AIP2, TOR1AIP1, CIITA, C12orf23, USP6NL, DHRS13, APOBEC3A, CCNI, TOPORS, EPM2AIP1, MLH1, FUT11, TIMM17B, PQBP1, SAA1, THOC7, ATXN7, SIRT2, ZCCHC3, PBXIP1, IRAK3, RAD51L3, FNDC8, TAP1, PSMB9, ISG20, ARHGAP24, PNRC1, DNAJC10, YARS, S100PBP, BTN3A2, RTP4, ELOVL5, DAXX, GAR1, LIMS2, GPR17, SERPINE1, YARS, S100PBP, ZNF644, POMP, RNF113A, NDUFA1, VWA5B2, CTCF, USHBP1, C19orf62, GAB1, C18orf56, TYMS, GABBR1, WDFY3, C4orf12, PRR5L, POLR3B, SPOP, VRK2, HABP4, NUP107, DLGAP2, ZNF566, C1orf59, TMEM127, CIAO1, RECQL4, LRRC14, HORMAD2, PIK3IP1, GLO1, CNR2, FAM71E1, C19orf63, TOR2A, BRWD1, SHE, TDRD10, CDC2L5, C21orf58, PCNT, MRE11A, ANKRD49, ERICH1, HLA-C, CHERP, TRIM9, CMTM2, CCDC62, HSPA8, SMC1B, RIBC2, PDE4DIP, ARHGAP25, USP49, GFI1, NOS1AP, UCP2, CLINT1, GGNBP2, RNF141, EXTL3, EXOSC9, SFT2D3, SEP15, HS2ST1, CACNA1I, ALG9, IKBIP, APAF1, SH2D4B, RAB33B, CLIP1, LOC344967, N4BP2, LOC100127888, LOC492303, ZNF326, FGFR4, HSPB11, LRRC42, HIST1H1C, ARF1, DHRS13, DSC3, SRGAP3, C6orf170, TAF6, CNPY4, MBLAC1, RAB37, CITED2, COG8, NIP7, NDUFA8, MORN5, SRP9, ZNF616, MTA3, FNBP4, GNGT2, ABI3, PELI3, ZNF423, CHDH, IL17RB, PPAT, PAICS, RASAL1, SRGAP3, CST8, ARF6, DIRAS2, LOC100128164, SEC62, MRPS36, FAIM, RAG2, C11orf74, USP50, GPR146, RAB3IL1, ARPP-21, HPS4, SRRD, UBE2J1, KRAS, C14orf142, UBR7, PTPRB, MAN1A2, VPS13B, MLL2, ZFP161, ZC3H7A, CD79B, EBF4, HNRNPK, RMI1, STRBP, OSGEP, APEX1, LOC100128164, SEC62, SMCHD1, SYNGR1, FLJ25006, LOC645851, SENP6, MAP1LC3A, C1orf156, C1orf112, CTHRC1, SCAP, C1QL4, ERCC6, ATPAF2, C17orf39, CALD1, PIK3C3, PAX6, WIPF1, COCH, KCNA3, MTMR10, RAB8B, CANT1, MANBA, POU5F1, RHBDL3, TIAL1, PIAS2, KPNA4  Figueroa et al., 201352 
SESN2, E2F6, RP11-363D14.1, AC131097.3, XIRP1, ZDHHC3, RP11-803B1.2, SIDT1, RP11-71E19.2*, APBB2, KIF2A, DUSP1, RP11-274H24.1, TBC1D7, ZNF90P2, CUX1, ERICH1, ERICH1-AS1, ZNF704, FARP1, TMED6, CBFA2T3, IGF2BP1, AXIN2, MIB1, DSC3, EPOR, TCFL5  Nordlund et al., 201553 
SOX11, SPSB1, BEST3, SIDT1, TCFL5, CHL1, FAM19A  Busche et al., 201359 
Table 4.

Methylation profile of the hyperdiploid subtype in pediatric ALL-B.

Hypermethylated
MYO3A, WWTR1, VIPR2, PID1, FAM20A, SOX11, CRMP1, KL, WWC1, FGFR2, ZNF532, SLIT3, ENPP5, LOX, B3GALNT1, KIAA1456, ADAMTS1, AJAP1, DYNLRB2, AJAP1, MFSD7, DPY19L3, MAGI1, NRG1, PTPRG, FOXL1, FBN2, DSC3, FHIT, DSC3, FBN2, BMPR1B  Chatterton et al., 201451 
PPAT, PAICS, KCNF1, HORMAD2, FAIM, APPL1, DUSP15, TTLL9, LCK, CLSTN2, TXNDC11, CSDE1, ASB8, SETBP1, COL4A1, COL4A2, TEAD1, C1orf183, HIST1H1C, OIP5, NUSAP1, SCAP, VPS13B, ARHGAP25, SLC39A1, CREB3L4, RPS13, ABCB10, DSC2, POMP, SLC35B3, PDLIM3, WBP11, C12orf60, ACSM3, SEP15, HS2ST1, SCAP, NPR1, FAT1, INHBE, GLI1, ASAP2, BEST3, LOC145783, TCF12, TFAP2C, CNKSR2, PHLDB2, FBN2, EXTL3, HK2, CDC14B, C11orf30, RELN, TNIP1, HPS4, SRRD, USP11, GFI1, HNRNPK, RMI1, APOBEC3A, CST8, PBX3, ACACA, TADA2A, OSGEP, APEX1, C14orf142, UBR7, LOC100128164, SEC62, SRBD1, HLA-DQA1, PAX6, RAD50, ETV7, CHMP4C, MYOD1, CALD1, CSDA, CNTN2, KPNA4, GFAP  Figueroa et al., 201352 
Hypomethylated   
LOC284837, TNXB, C14orf49, LOC284837, NKG7, KRT36, ENTPD1, APOLD1, LSP1, DBNDD1, CLDN16, C14orf49, SEPT9, CHST15, BIN2, BCL2, BCL9L, CEBPG, VTCN1, CTNND1, KANK2, SLAMF1, LGALS3BP  Chatterton et al., 201451 
MX2, C14orf49, C14orf49, ACTR3C, LRRC61, SYNE1, FAM195A, WDR90, ARD1B, BCL3, SYNE1, SERPINA3, SERPINA4, CEP350, DEFA1, DEFA1B, DEFA3, NPBWR2, LRRTM4, ARID3A, C21orf15, ZNF366, MMP26, KRTAP12-1, SERPINA10, PARK2, PACRG, CLCA2, ARID3A, SERPINA4, FAM90A20, DCAF4L2, RAB24, PRELID1, SYN2, SH2D3C, PLEK, TRPT1, NUDT22, GEMIN4, ELP2P, SERPINA10, PNMA3, GPR56, XKR5, NFATC3, BGN, IL13RA1, LOX, TNXB, MX2, GP2, BGN, MAGEC2, AMT, NICN1, KL, KLF6, FAM92A3, ZDHHC21, VTCN1, NRBP2, ABCA12, GPR84, CRIP1, C9orf66, DOCK8, UBXN11, SH3D20, BTBD16, DARC, CYP2F1, ATP2B3, E2F2, B3GAT3, COASY, MLX, MAGEA12, CSAG1, CCT8L2, MMP12, NDUFS8, CLEC4D, C21orf84, KCP, EIF2C2, S100A13, S100A1, C1orf77, CLEC4D, FAM47A, FES, CALML4, COL1A1, PLEKHJ1, NAPRT1, MAGIX, FGD5, C7orf26, IFFO1, NCRNA00095, FBXL19, C9orf116, MRPS2, DCI, C10orf11, SCRIB, PBXIP1, PYGO2, C21orf84, CDK5, SLC4A2, NCRNA00114, STRN4, FKRP, CD300LD, KLC1, CNGB3, DARC, ARHGAP30, NGB, TSEN2, DDAH2, KLHL17, PLEKHN1, RYBP, GEM, FUT11, C9orf79, CRYAA, GP1BB, NOLC1, KRT15, FAM151A, HRCT1, LOC158376, KDM4B, PNMA3, ADAMTSL3, THAP11, TPRXL, TGIF2LY, TGIF2LX, GPR162, COASY, MLX, KIF2B, KRT6C, DCI, KLK6, RFNG, GPS1, C14orf79, ZNF683, PTGDS, LCNL1, SIGLEC7, GPR39, YDJC, CCDC116, SSRP1, P2RX3, FAM189B, LOC642587, FAM195A, WDR90, PCDH12, KIAA1967, ACTRT2, CALML4, TMEM179B, TMEM223, DDAH2, ROBO1, LRRC16B, SLC29A3, S100A3, KRTAP12-1, TMPRSS3, LOC90834, ST3GAL6, SIGLEC9, CDK3, PLA2R1, RFPL3, TLR9, XAF1, POU5F1B, CHPF, TMEM198, IP6K3, CRAT, PPP2R4, HDAC1, BTBD16, MEFV, SIGLEC7, DNAJC4, VEGFB, CRYBB3, CCR7, DCST2, DCST1, STXBP6, ABCB6, C2orf24, FAM134A, ATF6B, FKBPL, RAB37, TMPRSS13, UGT3A2, HAUS7, LY6G6E, LY6G6D, C9orf69, FAM65A, CCDC12, NBEAL2, CCL25, ASGR1, RASSF1, ZMYND10, EIF6, LOC643008, ENTPD3, MEFV, INPP5E, ST6GALNAC4, KRT4, NPW, LIMD1, SLC26A6, SLC2A7, LCN10, LCN6, MAP3K8, SLC25A45, CCDC28B, GRINA, PSAPL1, CTRB2, CSF1R, NMUR1, STOML1, PML, CA5A, PRDX2, RNASEH2A, MUC4, SCARF2, KRT6A, CARD9, C6orf126, C6orf127, LOC401431, ATP6V0E2, PDE11A, C21orf129, NCRNA00112, GPR81, NEK10, SMTN, STXBP6, FCGR2A, GRINA, PCK1, EDNRA, FAM118A, C9orf79, LOC400940, GRM4, CAMKV, DEFA1, DEFA1B, DEFA3, ATF6B, FKBPL, KRT1, XYLT2, LYPD4, DMRTC2, S100A13, S100A1, C9orf79, ATP2B4, PALMD, LOC26102, GPSM1, FAM151A, SCNN1A, CDH5, SIGLEC15, SOX15, BACH1, POLD1, RAB11FIP1, VTCN1, MYEOV, GRIA3, EREG, HSPG2, FGF11, CHRNB1, DENND2D, RUNX1T1, C14orf70, PTH2, CACNG1, PRICKLE2, MUPCDH, SCT, LGALS7B, PTMS, LAG3, C8orf74, CLDN4, GNGT2, ABI3, KCNE1, C8ORFK29, FBXL6, GPR172A, UBE2L6, UCN2, MAGEB6, ZNF19, GRIA3, GSDMD, GNAZ, LOC401431, ATP6V0E2, C19orf59, TRAPPC5, B4GALNT2, NXF4, MEIS3, ATP4A, C9orf116, MRPS2, ARHGAP27C10orf116, AGAP11, ACD, PARD6A, C16orf48, C16orf86, CPNE7, SH3KBP1, SCARF2, CD79B, SNAI3, LCN10, LCN6, DGCR6L, SH2D3A, VAV1, STBD1, ADAMTSL2, EP400NL, FPGS, WFIKKN2, KLHL5, FXYD3, PIP4K2A, SLC35A2, SMPX, EGLN3, MICALL1, ID2B, S100A9, FTSJ2, NUDT1, ARHGAP24, SHC1, CKS1B, CYP2J2, GANAB, INTS5, MRI1, MFSD10, C4orf10, NAT9, TMEM104, C14orf139, SHARPIN, MAF1, KIAA1875, C19orf59, TRAPPC5, RGS7BP, E2F2, KCNE1, TLR1, DVL2, PHF23, PLCXD2, IRF8, ADM2, MIOX, EPHA2, SSTR2, MYO1A, LZTS1, FLJ90757, BAIAP2, NEDD9, FBXW5, C8G, PNLIPRP2, SPEM1, RXRB, SLC39A7, HSD17B8, DHRS13, SERPINE1, C4orf50, MYH7, MUPCDH, SCT, UBXN6, C17orf61, NLGN2, TCP11, CANT1, FBXO6, SLAMF8, CXCR3, FBXW5, C8G, DYNLT1, SYTL3, HSPG2, MTA2, PARS2, LTA, TNF, C9orf103, LEPREL2, CCDC96, TADA2B, CXorf22, AARSD1, RUNDC1, SOX15, EPHB1, ATP2A1, FASN, PLA2G2E, KIAA1598, C9orf69, APPL2, SP100, FHL2, ITGB2, TRPT1, NUDT22, DNAJC4, MAP6D1, BTBD1, CXCL1, RFTN1, CCL25, GPR126, AOC2, ICT1, CCL17, POU5F1, PSORS1C3, OTOS, PARK2, PACRG, PRKAG3, C1orf56, VPS29, RAD9B, NLRP10, NDN, ANO7, NCLN, PRICKLE3, CTSZ, WFIKKN2, AK1, TMEM37, TUBB2A, KCTD11, TMEM95, KCNK18, CABP1, ANO4, RLN3, IL27RA, SSTR1, EIF6, BRMS1, B3GNT1, ERCC6, DHRS13, C2orf90, VWA3B, GUK1, LOC100240726, ENPP6, HOMER3, NLRP10, COMTD1, CIZ1, DNM1, IL25, CMTM5, CCR6, MARCKSL1, SHARPIN, MAF1, KIAA1875, IL11RA, C1orf231, MMACHC, KCNAB3, TRAPPC1, CNTROB, B3GNT5, ANGPT4, LOC100129637, DEFB124, REM1, PIGZ, FAM96B, CES2, ARMC3, ADRA1D, PTGS1, OR1L4, DAPK2, CMTM2, KCNAB3, TRAPPC1, CNTROB, RAB4A, PRKCH, GPR126, BBC3, CUEDC1, PHLDB1, MAGEB6, HABP4, DLGAP2, TLR2  Figueroa et al., 201352 
S100A16, HNRPLL, DNAJB2, SH3BP5, LARS2, RGS12, RBPJ, DDIT4L, ANKRD33B,RP1-209A6.1, TNXB, CLEC5A, PKIA, EIF2C2, LCN6, C9orf139;FUT7, RP11-393K12.2, MARCH8, ALDH3B2, RAD51L1, SYNE3, SEZ6, MOBKL2A, PLVAP, CLDN14, LOC284837, CELSR1  Nordlund et al., 201553 
Table 5.

Methylation profile of other subtypes of pediatric ALL-B.

Subtype  Methylation profile  Reference 
Translocated MLL, BCR-ABL and TCF3-PBX1Hypermethylated genes:WWC1, NELL1, SEMA3C, FGFR2, ARNT2, NRG1, FBN2, KCNC3, SSH3, DSC3, CYB5R2  Chatterton et al., 201451
Hypomethylated genes:CHST12, LSP1 
Translocated MLLHypermethylated genes:ZAP70, XYLT2, HLA-B, EDEM1, UBXN11, CD52, C14orf43, HLA-C, MICALL1, C14orf43, PRKCH, TP53I11, GHRL, SNORD10, SNORA67, CD68, MPDU1, ITIH3, CMTM2, PTPRE, MOV10, GUK1, LOC100128164, SEC62, IFITM2, SELO, TOR2A, C3orf37, CD79B, VWA5B2, CD74, C22orf26, LOC150381, ARF1, SORBS3, APPL2, AK1, CCDC102A, GPR114, KPTN, PPP1R14B, PLCB3, KCNK3, LRRC15, ITPRIP, GAB1, PPP1R14B, PLCB3, GP9, CACNB3, FOXN3, FDX1L, AFF3, SNORA77, PRX, CLINT1, UCKL1, UCKL1AS, CAMTA2, FTSJ2, NUDT1, TMEM220, DLGAP2, AMIGO3, GMPPB, KPTN, ANKRD13D, NINJ1, SSTR2, PPP1CA, TBC1D10C, SIK1, SBSN, GAPDHS, PRKCB, IL2RA, KLHDC7A, DEGS1, LIMD1, AK1, CTCF, ZNF267, CCR6, ANGPT4, DENND3, LTB, LST1, MDC1, TUBB, FLJ42393, MCF2L, C1R, HPCAL1, C21orf84, SPAG8, HINT2, SLC19A1, PLA2G4D, NLRP12, GUK1, PRSS27, LSM7, SPPL2B, GADD45B, TMEM220, TMEM204, ANKRD13D, CTDSP2, SH2D4B, STK32C, EFNA3, ANGPT4, SPIB, IRAK3, NDUFB11, RBM10, APP, ARHGAP27, TMEM129, TACC3, SMTN, SPAG8, HINT2, DNAJC10, S100A13, S100A1, NUP107, HIRA, MRPL40, TENC1, OAF, C7orf61, DBNDD1, RPS11, SNORD35B, HSPA8, TAP1, PSMB9, RPAP1, REPIN1, NUDT8, DNTT, STRN4, FKRP, LTA, TNF, TMCO4, HABP4, NCK2, SSTR2, USP50, CXCR5, GYPC, XYLT2, B3GAT3, SIRT2, ARHGAP24, S100A13, S100A1, BCL7C, CTF1, COG8, NIP7, PTPN18, ZFP161,SLC27A2, PPP1R3E, BCL2L2, ITIH3, LOC401431, ATP6V0E2, STK32C, RAB37, MYO7A, GUK1, GJC2, PCNA, CDS2, STK38, HSPB11, LRRC42, STAB1, GP9, NCLN, RGS14, C21orf84, SERPINE1, COG3, CPNE7, BTK, RPL36A, SGPL1, MRPS15, PIK3CG, CRAT, PPP2R4, MDM4, IL3, MLL2, C1orf231, MMACHC, LOC401431, ATP6V0E2, CD22, FFAR1, MAP6D1, BCL7C, CTF1, TMEM127, CIAO1, CNR2, PXK, STX17, ADCK4, ITPKC, CPNE7, CD79B, SHARPIN, MAF1, KIAA1875, IL3, PELI3, SIGLEC15, C3orf42, GHRLOS, NUP205, MAP6D1, PRDM8, DHRS13, FASN, MUPCDH, SCT, LOC100127888, GRINA, RFTN1, IP6K1, AKAP8L, NCRNA00095, FBXL19, MTHFR, CLCN6, C22orf26, LOC150381, PIP4K2A, LOC100128164, SEC62, DAXX, CYFIP2, TNF, LTB, GANAB, INTS5, CLDND2, NKG7, CD37, C12orf49,RNFT2, POU2AF1, SOX15, ZNF366, TMEM127, CIAO1, SFN, LOC100127888, KRTAP5-8, SUV39H2, FAM189B, TIMM17B, PQBP1, ANKS6, TERT, KDM4B, C9orf116, MRPS2, GRINA, ZNRF3, POMP, MAG, MYL4, RHOH, DNAJC10, UGT3A2, BTN3A2, C22orf24, YWHAH, ZNF566, TINAG, ZFHX3, PBXIP1, TGM6, S100A13, S100A1, ACTRT2, ALPPL2, WIPF1, RECQL4, LRRC14, NCKIPSD, R3HCC1, RAB24, PRELID1, SOX15, LY6G6E, LY6G6D, HPS4, SRRD, CANT1, ADM2, MIOX, ATP2B3, SNORA67, CD68, MPDU1, FLJ25006, LOC645851, C21orf129, NCRNA00112, LEPREL2, LTB, LST1, GPER, DHRS13, MUC4, COG3, GNB4, RANBP9, DCUN1D2, TMCO3, RAB33B, ST6GALNAC3, C1QTNF4, STBD1, POLD1, LRRC6, GFAP, LRFN2, MTMR10, FITM1, PSME1, SFT2D1, CBLN3, KHNYN, DAPK3, HBEGF, SIDT1, FUCA1, CSDA, C14orf49, VKORC1, CISH, MAPKAPK3, RAB11FIP1, GFAP, KRTAP12-1, ITSN2, CIZ1, DNM1, TMED6, MAN2A2, MAP1LC3A, SORL1, RAG2, C11orf74, TIMM13, KLHDC5  Figueroa et al., 201352 
CDH3, TBX2, ERCC1, NPR2, DAPK1, CCR6, HRK, LIFR1, DLX3, FHIT  Stumpel et al., 200960; Schafer et al., 201061 
Hypomethylated genes:GUCY1A3, JMJD1C, LOC84989, BAZ2B, PRF1, RUNX3, PRF1, MAP7, RHOBTB3, RUNX3, LARGE, SLC25A18, ITGAE, FLT3, NPR1, FAM65B, PTGR1, ACSL1, MEF2C, TRPV3, SKA1, CNKSR2, H1F0, GCAT, F3, MTHFD2L, TLR7, ARRDC4, DAPK1, PSMB5, TMEM41B, LOC100303728, SLC25A5, ZNF367, FLT3, LOH12CR2, LOH12CR1, PHLPP1, SERPINB13, OR6V1, PRDM11, CD3D, CD3G, SENP6, TRAK2, STRADB, ASTN2, TRIM32, SETBP1, IGF2BP2, TFCP2L1, CENPF, MPP5, C7orf13, RNF32, PSMB5, C7orf13, RNF32, CD180, PARP8, RBKS, LOC100302650, BRE, ARID1B, C6orf122, C4orf19, MGC16703, P2RX6, OLFML3, IQSEC1, MYST2, KRTAP10-6, PDPN, NID1, CPD, LCE3A, SSR1, CDKL3, UBE2B, PAPOLA, ANKRD27, RGS9BP, HTR7, DES, FAM5B, CDC14B, GPR153, SPOCK2, ITGAE, LECT2, SERPINB8, ZAR1L, BRCA2, CAMTA1, C9orf100, ALDH4A1, C17orf64, KRT81, LILRB4, MRPS25, ASB8, SLC25A38, TRAS, MYOD1, DKK3, PPP1R3B, BDNF, CCBL1, LRRC8A, RASGRF2, GRIK5, KCNJ10, FAIM, MTMR11, PEA15, SBF2, HECW1, WDYHV1, PHLDB2, GINS3, PDE4DIP, KDM5B, ZNF256, PHLDB1  Figueroa et al., 201352 
ZSCAN18, ZNF256, ZNF329, ZNF544, ZNF681  Nordlund et al., 201353 
BCR-ABLHypermethylated genes:KIAA1949, NRM, MUPCDH, SCT, ATP2A1, FASN, DPM2, PGLS, MYEOV, DUSP5, MARCKSL1, ANKRD27, RGS9BP, ADCK5, FOXH1, C11orf16, DIRAS1, MRPL24, DCST2, DCST1, CCDC71, INPP5E, SLC22A20, GPR146, PLEKHM2, PROC, DIRAS1, ALDH4A1, WFIKKN2, IL11, LIMS2, GPR17, GRINA, LILRB4, C19orf59, TRAPPC5, CD38, LOC100129637, PRPH, LOXL4, KLHL24, ARHGAP27, GPR146, SSR1, C19orf59, TRAPPC5, C17orf61, NLGN2, C9orf116, MRPS2, DLG4, ACADVL, TLR9, STOML1, PML, KLF13, KCNK17, KLC1, ARHGAP30, C8ORFK29, FBXL6, GPR172A, DLG4, ACADVL, TAF6, CNPY4, MBLAC1, C9orf69, AURKC, PCOLCE, PTMS, LAG3, DNM1P35, GRINA, BSCL2, GNG3, CCL25, TEX19, CDH5, TLR9, CDK5, SLC4A2, GUK1, C9orf116, MRPS2, S100A13, S100A1, FXYD2, GPR153, LOC100129637, FLJ90757, BAIAP2, IP6K3, KLF6, LPAR2, UCN2, KCTD11,TMEM95, TFR2, C14orf70, CARHSP1, KLF13, CCDC96, TADA2B, LIMS2, GPR17, COL1A1, ARID3A, SHARPIN,MAF1, KIAA1875, SIRT2, WFIKKN2, ZNF296, GEMIN7, RFNG, GPS1, SLC26A8, MAPK14, FAM176B, CCBL1, LRRC8A, KCNK17, KCNAB3, TRAPPC1, CNTROB, MAGIX, PCDH12, SHARPIN, MAF1, KIAA1875, C16orf59, C9orf69, SP100, LACE1, C17orf64, CSF1R, MUC4, C1QL4, KCNAB3, TRAPPC1, CNTROB, GRB7, CLDN4, CARD9, ATP1A3, C1orf220, GUK1, FAM109B, C22orf32, CLTB, HSF2BP, RRP1B, GABBR1, B3GNT5, ITIH3, LOC26102, GPSM1, UBXN6, SH3D20, SIGLEC7, EDC4, CCR7, ACTRT2, CTXN1, RAB3IL1, DNAJC4, VEGFB, ADM2, MIOX, TUBB2A, SSX2IP, C16orf81, LIMS2, GPR17, ZNF48, PKNOX2, GRASP, PECR, TMEM169, NTN5, C9orf103, CACNA1I, BTG2, ADRA1D, DHRS13, THEM4, MT1L, PHLDB2, PDE4DIP, IL25, CMTM5, LIF, KLHDC5, PITPNM3, GALP, FAM109B, C22orf32, PIGZ, PRDM11, CNTN2, MIB1, BSN, MOCOS, DRD5, COL1A2, KIAA1257, LYPD4, DMRTC2, POU5F1, RASL11B, GFAP  Figueroa et al., 201352 
Hypomethylated genes:GBA2, RGP1, DPP3, C11orf30, ECHDC1, ZEB2, INHBE, GLI1, INTU, WBP11, C12orf60, STARD3NL, C21orf59, FGF6, ITGA2, FBXW7, OIP5, NUSAP1, C6orf47, BAT4, CSNK2B, RPS13, RPL4, SNORD18C, SNORD18B, SNORD16, SNORD18A, ZWILCH, CCDC115, IMP4, POLR1E, C22orf26, LOC150381, ANG, RNASE4, SRBD1, GIMAP5, C1orf183, SLC25A28, TRIB3, CTNNA2, STBD1, C22orf26, LOC150381, IQGAP2, RHBDD3, EWSR1, CSDA, C3orf42, GHRLOS, CYP1B1, GPR6, MFI2, TTC23L, OGFRL1, LRRTM1, CCDC70, C5orf43, NKX2-1, ADAMTS12, NDUFC1, NARG1, TUBA4A, TUBA4B, CLIC2, TMEFF2, KIAA0087, UGGT2, CACNA1B, PRPF8, PJA2, CXCL12, SLC20A1, ACSBG1, CA6, PDZRN3, RTDR1, RAB36, ROBO4, BRDT, PROX1, COL14A1, FLJ14107, ATOH1, NLGN4X, SLC35A1, LRIG3, LANCL1, CPS1, HOXD1, KCNN2, PCNA, CDS2, IL2RA, ZNF264, PCDHGB7, PCDHGA11, GAB1, FGF5, SALL3, CISH, MAPKAPK3, C15orf24, PGBD4, SFN, RIC3, FAM190A, LIMD1, MTMR11, EPHA5, IRX2,C5orf38, ZNF804A, HIPK2, TBXAS1, TINAG, BPNT1, IARS2, MSX2, GLIPR1L2, PRDM8, BDNF, MTERFD2, HOXD1, LRFN5, CDCA5, ZFPL1, MAL2, CDH8, CD22, PCDHB18, PCDHB19P, KCNK13, PKD2L2, GZF1, PDPN, VKORC1, DAXX, FOXQ1, TNFRSF9, MRPL1, TRIM13, UCHL5, TROVE2, EFR3A, SLC39A1, CREB3L4, WIPF1, XYLT2, CPVL, RAG2, C11orf74, PCDHB7, PCDHB8, BCL2L13, METTL8, DCAF17, LRP11, CHMP4C  Figueroa et al., 201352 
TCF3-PBX1Hypermethylated genes:FNBP4, CLDND2, NKG7, ARPP-21, SOCS2, NCK2, ARHGEF6, GNA13, TLR1, TMEM156, CD79B, HABP4, ISG20, FOXM1, C12orf32, LAIR1, HECA, CST4, STX17, NFKBIE, TMEM151B, PIGV, MLKL, FDXACB1, C11orf1, ASGR2, EFHC2, GNGT2, ABI3, USHBP1, C19orf62, ZNF25, OAS3, CYSLTR1, GAB1, GNGT2, ABI3, SORBS3, KRAS, BEST1, ARAP3, POLD1, HSPB11, LRRC42, RAB37, ITPRIP, MTA3, KIAA2013, SERINC5, ARHGAP24, KLHL5, CCL25, IL2RA, B3GNT9, TRADD, FBXL8, UBE2L6, KLHL21, GABBR1, OLFML3, NGF, C5orf56, PCNA, CDS2, SSRP1, P2RX3, TMEM127, CIAO1, STAB1, ETFA, TSFM, LOC100129066, ZNF683, ARHGAP24, HSD17B4, CPNE8, NUP107, UGT3A2, FBXO6, CLEC4D, VRK2, C11orf75, PDK1, CCL17, PSTPIP1, CEP350, IKBIP, APAF1, VWA3B, HGF, TMEM217, TBC1D22B, C17orf64, RNF113A, NDUFA1, HELZ, BTG2, SMARCB1, EIF4E3, GPR27, MEF2C, DUSP4, CTDSP2, NUDT5, CDC123, ATP5G2, ST8SIA1, PRDM8, CTCF, SMTN, CMTM2, DAXX, KLHL24, CACNA1I, NCLN, PTPLAD2, ERCC6, HBEGF, PRDX1, FCGR2A, CLU, TNFAIP8, LTB, LST1, FOXN3, MCM3AP, C21orf57, SMC1B, RIBC2, BTG2, SMC6, GEN1, DAZAP2, SYNGR1, CDC2L5, LMAN2, UTP6, C22orf24, YWHAH, SIGLEC15, LOC646999, STRA6, NOLC1, NCAPH, SENP6, ZNF423, NCKIPSD, PPP1R15B, SERPINE1, KPTN, FAM60A, FLJ13224, LDLRAD3, LCN10, LCN6, SLC38A5, FTSJ1, SPOCD1, FPGS, ANKS6, WIPF1, MTMR11, MRPS15, NOLC1, C20orf197, ZNF652, DPY19L3, ATP2B4, SRGAP3, EN1, ZNF41, CYBRD1, RANBP9, LRP11, LTB, LST1  Figueroa et al., 201352 
Hypomethylated genes:ZNF512, CRYM, WAPAL, SLAMF1, MYBPH, EXTL3, FAM109B, C22orf32, LHCGR, AKR1B1, SURF1, SURF2, PLXNC1, WDR87, SIPA1L3, NOL11, EXTL3, SDF4, B3GALT6, DEF6, NCRNA00176, LIPC, NFKB1, ASB8, TERT, NSUN4, LOC100129534, ADIPOQ, PHYHD1, BLK, LPAR2, FAM109B, C22orf32, GJA4, ADIPOQ, PDCD1, MND1, RASL11B, RNF186, PSMC3IP, MRPL28, ACTL9, TNS4, LAT, FAM109B, C22orf32, TEKT2, ADPRHL2, NAGA, FAM109B, C22orf32, PLD4, TEKT2, ADPRHL2, C16orf11, GLYCTK, NGF, PIP4K2B, ACOXL, MAST4, AURKC, SOST, MAP6D1, POLM, MSRB3, VAMP2, SLC39A1, CREB3L4, IDH2, NAT1, SSR1, ST6GAL2, FXYD2, ST6GALNAC3, CDCA3, USP5, PHACTR3, FFAR1, SDF4, B3GALT6, GDPD5, ARL4C, ATXN7L2, FAT1, GLYCTK, TRIM67, CTBP1, C4orf42, ARL4C, HOXD1, TSPAN3, TBX6, YPEL3, B3GNT7, MUPCDH, SCT, HIST1H1C, ITIH3, C12orf53, SOX21, SALL3, HECW1, PEA15, MNAT1, WNT2, PPARD, C16orf81, LILRB4, WNT2, C9orf100, SOX9, NCRNA00176, TFAP2C, C16orf81, GPR123, HOXD1, NXPH2, USP11, LTA, TNF, SCAP  Figueroa et al., 201352 
Rearranged CRLF2Hypermethylated genes:STXBP5, SLC2A8, GFRA1, DSC3, C9orf150, LCE3A, CHRDL1, C15orf24, PGBD4, HOXB2, ST6GAL2, NOVA1, NOL4, SLITRK1, TMSB15A, PRPF19, LRRC57, HAUS2, C18orf34, EPHA5, DSC2, RBPMS, TUBGCP3, AQP4, C18orf16, GABRQ, TMTC1, AADAT, ZIC5, MYEOV, ASXL1, NEUROG1, EPHA5, SIX6, TMED3  Figueroa et al., 201352 
Hypomethylated genes:HIPO, LYSMD4, SRBD1, PYGM, CASQ1, KCTD3, ANO7, WIPF1, CA6, GP1BB, NLRX1, LCN10, LCN6, SLC2A5, S100A13, S100A1   
Altered ERGHypermethylated genes:CDKN2C, CYSLTR1, TMEM156, PIGV, RFTN1, SOCS2, MTHFS, MLKL, SERINC5,CD48, FLJ14107, ENAH, TNF, LTB, DAXX, RFTN1, TNFAIP8, TUBA4A, TUBA4B, HIBCH, NUBPL, C9orf25, DNAI1, STIM2, CHMP1B, PEX16, GYLTL1B, TLR1, CH25H, DEGS1, C8orf56, BAALC, XPO5, POLH, CXCR3, MED14, DDX3X, DENND3, ZNRF3, PELI3, PIK3IP1, HELZ, TGM6, GAB1, RBP1, C1QL4, ATP2B4, PRR5L, SLC27A2, ATP10A, LTB, LST1, STIM2, TMEM178, BNIP3L, TNFAIP8, CEP350, ZMYND8, UBE2J1, CLINT1, EXOSC9, MTMR10, FPGS, CCNI, CDKN1A, UBXN11, FAM120AOS,FAM120A, SYNGR1, ITGB2, RNF141, CD3D, CD3G, SRGAP3, CHRNA2, ZNF490, ZNF791, TP53BP1, WBP11,C12orf60, WDFY3, C4orf12, CARD8, GGNBP2, IKBKE, UTP20, RAPGEF1, HSPA8, PPP1R15B, SENP6, ST6GALNAC3, CD48, KDM4B, PPID, ERO1LB, SH2D3A, VAV1, KCTD17, ZNF212, ARHGAP24, RANBP10, TSNAXIP1, CYTIP, LTB, LST1, LOC729234, PKNOX2, C8orf56, BAALC, SYNE1, DYNLT1, SYTL3, TRIM13, UBR5, IGFBP7, ERAP2, CDCA5, ZFPL1, PARS2, MEF2C, PIP4K2A, NLRP12, ANKFY1, ST3GAL6, PJA2, UGT3A2, DHRS13, FUCA1, CENPJ, RGMA, RAB39, LRRC8D, RANBP9, AIF1, ITPRIP, TAF3, ERCC6, MRPS15, SRGAP3, CDCA3, USP5, NGRN, PIP4K2A, L1CAM, SERPINE1, EFHD2, FXYD5, NUP107, NARG1L, MAP3K8, C14orf33, KTN1, RPRML, RNF113A, NDUFA1, ADAMTS5, MDM4, TLR7, EGLN3, HYAL3, NAT6, HYAL1, NOVA1, DCTN2, KIF5A, TRIM9, ST6GALNAC1, C1orf127, WIPF1, LRRC57, HAUS2, CYBRD1, SLAMF1, ACOXL, PBK, ECM1, RASL11B, TMEM37, GRPEL2, ACSL3, ICAM3, C1orf156, C1orf112, PXT1, KCTD20, STXBP6, TBCCD1, DNAJB11, SIDT1, FGFR4, SLC16A9, CAB39L, SETDB2, ERICH1, DTX3, SSTR2, CBX6, VRK2, C14orf48, EIF4E3, GPR27, SMCHD1, XYLT2, TSEN2, CEP70, STXBP6, USHBP1, C19orf62, ST6GALNAC2, ADAP2, GLRA2, WFDC2, NOLC1, GFI1, CD180, NP, SLC39A13, IRAK3, SPRYD5, C9orf79, DNAJC9, MRPS16, LYPD5, ZNF283, TMEM217, TBC1D22B, ENTPD3, HSPG2, CCDC85A, STK38, CNR2, KIR3DX1, IL8RA, CECR2, MYL12A, MYST2, KCNAB1, WDR47, PYY, NAGS, GNAZ, DEFA1, DEFA1B, DEFA3, C17orf64, RPL17, SNORD58C, U58, SNORD58A, SNORD58B, ESPL1, PXK, CARD14, HNF4A, C9orf79, PCDHB2, CD33, DGKG, S100A13, S100A1, FIBIN, ATP5G2, RAB11FIP1, GREB1L, GJA4, PRICKLE2, RAG2, C11orf74, ZCCHC9, ZNF683, CXXC5, HNMT, C12orf23, ACSBG1, CLU, BPNT1, IARS2, CCR7, ALX3, GABRR1, TFB2M, C1orf71, DDX19A, CACNA1S, THYN1, ACAD8, PTH2, KIF12, GNGT2, ABI3, CAMK2B, SEMA4A, ADARB2, MC4R, DNAJC10, TMPRSS13, C7orf43, DOCK3, DCK, NBLA00301, STBD1, USP2, CDR2, SSRP1,P2RX3, IKBIP, APAF1, PNPT1, DCAF5, ZNF423, PLEKHH3, CCR10, CNTNAP1, LEMD1, PCDHB7, PCDHB8, MLL2, XPNPEP2, DYNC2H1, CPOX, PIAS2, GMNN, HMP19, SEL1L3, PTPRB, CRYBB3, C14orf115, NELL1, ITIH5, RRAGA, LRAT, DEFB124, REM1, C15orf24, PGBD4, B4GALT3, RHBDL3, SRBD1, HTR7, FAM193B, GNB1L, C22orf29, AFAP1, BRDT, MRPL1, DIAPH2, PRICKLE3, ACTRT1, TNFRSF9, SSTR2, HNF4A, SOCS3, SLC39A1, CREB3L4, KCNAB3, TRAPPC1, CNTROB, FOXO4, NOV, NOLC1, DNAJB1, C9orf79, LOC400940, TMPRSS2, GPR84, ZNF366, SFT2D3, TUSC3, VPS72, DCBLD1, AMMECR1L, SLC2A14, PRR18, VPS24, DNAJC10, EXOC8, C1orf124, MAL2, TPRXL, YARS, S100PBP, PO   
Hypomethylated genes:A2LD1, SAP18, BTN2A1, OSBPL11, HN1, AARS2, ADI1, OBFC2B, SLC39A5, ZNF295, PARVB, MIA, RAB4B, ZNHIT2, FAU, MRPL49, TULP4, C7orf65, TMIE, PSMA2, MRPL32, SPINK4, ZNF296, GEMIN7, PEX11A, WDR93, RASSF2, PNRC1, SLC39A11, GIT2, ANKRD13A, DIXDC1, VPS4B, PARVB, CHRNA1, KRTAP5-8, ADAMTS9, CNTN2, C14orf139, EPS8, PCNX, LDOC1L, CCDC109A, PRMT10, CCNT1, NSD1, WDFY4, DCP1B, GCHFR, C12orf36, RFC4, TFF3, GPR81, ZNHIT6, C19orf23, C19orf24, PIK3R1, C16orf11, NHLRC4, PIGQ, C10orf118, TDRD1, XPNPEP2, SECISBP2, GLYCTK, CUL7, MRPL2, KLC4, GLG1, C7orf25, GPR153, EIF1AD, BANF1, PPL, TMEM127, CIAO1, GPR146, CAND2, DIXDC1, UCKL1, UCKL1AS, SNHG7, SNORA43, SNORA17, RFC4, NME3, MRPS34, EME2, ING1, USP12, HNRNPR, RGN, NHLRC4, PIGQ, TSKU, GRB2, CST8, SCGB3A1, ZNF513, ADCY1, LRRN1, C3orf27, EXOSC10, LFNG, XYLT1, C14orf126, GPR44, CRBN, C16orf81, GLIPR1L2, LTA4H, SPIB, CCNJ, GPR146, LOC643008, GLIPR1L2, BACH2, CCDC109A, GPR155, ZC3H18, ELP3, ALPPL2, CRBN, ADAMTS3, POLM, MRPL28, HEATR5B, CCDC75, GPER, CUL7, MRPL2, KLC4, CHRNA1, C14orf139, SREBF1, MRPL55, KCNC3, BTG2, MTSS1L, OR2Z1, LOC100303728, SLC25A5, CACNA1I, IFI30, GRINA, CRHBP, RNASE7, CD81, SURF1, SURF2, PTOV1, TGFBR2, TEKT2, ADPRHL2, ADAMTS3, IGSF21, SOHLH1, KCNT1, RPL9, LIAS, SHARPIN, MAF1, KIAA1875, GRINA, C20orf165, NEURL2, CTSA, SLC20A2, C8orf40, SNW1, C14orf178, RHBDF1, MPG, LOC26102, GPSM1, EIF4G2, RHBDF1, MPG, TPI1, GLYCTK, ATP2A3, ARHGEF4, TBC1D13, CD38, MS4A10, LOC100129637, SOX15, LIF, C19orf23, CIRBP, C19orf24, ABCA2, C9orf139, CCDC96, TADA2B, BSCL2, GNG3, NELL1, RGS9, GPR148, GANAB, INTS5, HIST1H1C, BMP7, LOC100129637, CGREF1, ABHD1, PCP2, STXBP2, LOC643008, CIZ1, DNM1, GPER, TSKU, TRIM67, GFRA1, FLJ90757, BAIAP2, FTSJ2, NUDT1, DDX50, FOXJ2, SLCO3A1, LIN7A, GATM, RAMP1, COG8, NIP7, NFS1, ROMO1, VPS37C, FAM53B, G0S2, SHANK1, SPATA24, C19orf23, CIRBP, ZNF566, FAM83H, IMMP2L, LRRC3, TSTA3, HLF, SDF4, B3GALT6, LHCGR, C16orf42, GNPTG, SLC2A8, HS6ST1, C20orf165, NEURL2, CTSA, SFT2D1, CSMD1, RAD54L, PITRM1, MUPCDH, SCT, TIGD3, VAT1L, TOMM40, CCDC42, WARS, WDR25, GRIK5, CYP2U1, ODF3L2, HMP19, KLHL22, ARID3B, MT3, SOX15, DMRT3, REPIN1, MTA2, ATPAF2, C17orf39, SHARPIN, MAF1, KIAA1875, SQLE, CHMP4C, SYT15, MYH6, SNTB1, MRPL24, GPR123, GATA6, FBXW5, C8G, EPHA5, PRELID2, IVNS1ABP, KRT80, SNAI2, RBM3, VWDE, NUDT10, ZMYM6, MRPS30, TIMM13, ZNF532, SYVN1, NKX2-8, KIAA1024, FAM81A, KCNN2, MUC1, THYN1, ACAD8, LOC100130557, NFYC, ALOX5, CCDC58, FAM162A   

Figueroa et al. evaluated patients with ALL with both immunophenotypes. In their study, included 137 children with ALL-B and 30 with ALL-T and identified a signature of 85 genes with alterations in methylation, which correlated with the expression analysis (Table 2). The identified signature was common between all ALL subtypes (B and T) and included genes TIE1, MOS, CAMLG, GPRC5C, which are involved in signaling; MCTS1 and DGKG, which participate in regulation of the cell cycle and cell proliferation; PABPN1 and PABPC5, which participate in RNA metabolism; PROP1, TAF3, H2AFY2, ELF5, ZBTB16, CNOT1 and TADA2A, involved in transcriptional regulation, as well as homeotic genes HOXA5 and HOXA6.52 Additionally, this study determined specific methylation and expression signatures characteristic to every ALL-B subtype in children. Seven independent ALL-B groups that presented a characteristic methylation/expression signature, depending on the molecular rearrangement present in each group of patients were identified: rearranged CRFL2, ETV6-RUNX1, high hyperdiploidy, translocated MLL, altered ERG, BCR-ABL1 and TCF3-PBX1 (Tables 3-5).

The study by Norlund et al., which included a greater number of patients with pediatric ALL (663 children with ALL-B and 101 children with ALL-T), confirmed that each ALL subtype presents with a specific methylation signature. Based on this study, it was proposed that methylation signatures could be useful to predict the genetic subtype of pediatric ALL. This proposal would be applicable to cases in which difficulties arise with the identification of a chromosomal or molecular alteration that contributes to leukemia classification.53 Despite methylation signatures to determine ALL subtype have been found and epigenetic profiles to allow the prediction of the risk of relapse have been proposed,47,54 an epigenetic profile capable of accurately classifying and stratifying patients with ALL does not exist.48

3.2Alteration in gene methylation in ALL according to the specific subtype of genetic rearrangement

Besides alterations in DNA methylation, which are common to all subtypes of ALL-B, exclusive modifications to each genetic subtype exist.48,51,55 Recently, methylation profiles together with gene expression profiles in patients with the ETV6-RUNX1 fusion were analyzed; 55 genes with alterations in methylation were identified (Table 3).51 Among these alterations, EPOR gene was identified, which was found to be overexpressed, and its promoter, hypomethylated. The ETV6-RUNX1 fusion binds and activates the transcription of EPOR (erythropoietin receptor), which contributes to leukemia development, activating cellular proliferation and survival through the JAK2-STAT5 pathway.56 Hypomethylation in EPOR favors its promoter to be more permissive to binding and activation by ETV6-RUNX1.

In the ETV6-RUNX1 subtype, hypermethylation and low expression in the asparaginase synthetase (ASNS), which sensitizes cells to treatment with L-asparaginase was also found. This explains, in part, the fact that patients with ETV6-RUNX1 present such a good treatment response.57,58 Other genes with alteration in methylation and expression observed in ETV6-RUNX1-positive patients include CLIC5, ACVR1C, IGF2BP1, DSC2, PCLO, SOX11, SPSB1, BEST3, SIDT1, TCFL5, CHL1 and FAM19A (Table 3).55,59 Additionally, it has been reported that alterations in the methylation of ERHV-3, DMNBP, KCNA3, PAG1 and C11orf52 are associated with risk of relapse in patients with ETV6-RUNX1, for which they have been proposed as markers of poor prognosis.47

In patients with hyperdiploidy, alterations in methylation and gene expression that are exclusive to this subtype have been found. More recent studies highlight the hypermethylation and low expression in tumor suppressor genes FHIT, PTPRG and DDIT4L.47,51 Additionally, hypomethylation of proaptotic genes BCL2 and BCL9L has been observed, which have been reported to be altered in diverse B cell tumors (Table 4).51 On the other hand, the ALL subtype positive to rearrangements in the MLL gene also presents a characteristic profile of alteration in methylation (Table 5). It has been reported that in ALL that with rearrangement in MLL, hypermethylation of CDH3, TBX2, ERCC1, NPR2, DAPK1, CCR6, HRK, LIFR1, DLX3 and FHIT genes occurs.60,61 Hypermethylation of genes ZSCAN18, ZNF256, ZNF329, ZNF544 and ZNF681 has been associated with an increase in the risk of relapse in patients with alterations in the MLL gene.47 In particular, it has been observed that a different methylation profile is present in ALL with different translocations in MLL.60

3.3Main cellular processes and signaling pathways affected by alterations in gene methylation in ALL

Cell processes with a greater representation of genes with aberrant methylation in ALL include the regulation of the cell cycle, apoptosis, transcriptional regulation and cellular adhesion (Figure 3). Some examples of genes with alterations in methylation in ALL that participate in these cellular functions are CDKN1A, CDKN1C, CDKN2A, CDKN2B, SFRP, CDK6, which regulate cell proliferation and cell cycle62–66; GATA4, HLF, TCF3 FOXF2, PAX2, PAX5, PAX6, TFAP2A, TWIST72, TP73, HOXA4, HOXA5 and HOXA6, which participate in gene transcription 51,62,67–70; DAPK1, APC, EPHA2, RASSF1, PTEN, APAF1, DBC1, DCC, EPHA2, EYA2, miRNA34, miRNA34C, NOXA, ASPP1, PTPN6, RASSF6, SFRP1, WDR35, PAR4, ERBB4, involved in apoptosis4,20,36,51,71–73. Alterations in the methylation of these genes confer leukemic cells with survival advantages, preventing apoptosis and promoting proliferation (Figure 4).

Figure 3.

Classification of the most affected genes by epigenetic modifications in ALL, according to their biological function.

(0.25MB).
Figure 4.

Signaling pathways and cell processes that are modified by epigenetic alterations, which promote leukemogenesis.

(0.52MB).

Some signaling pathways mostly represented by genes with alterations in methylation in ALL are p53, WNT, EPHR, MAPK, and PI3K-AKT4,20,59, which are tightly related to cell processes as the aforementioned (Figure 4). One of the most profoundly studied pathway is the TP53, which is a tumor suppressor gene involved in the activation of damaged DNA repair, induction in the G1/S checkpoint and promotion of apoptosis.74 Despite mutations in this gene are present in 50% of tumors, less than 3% is found in ALL. However, an alteration of the p53 pathway at the epigenetic level exists in ALL.59 Hypermethylation of at least one gene involved in the p53 pathway has been observed in 78% of patients with ALL. Hypermethylation in genes involved in apoptosis dependent on p53, such as AIFM2, APAF1, DBC1, miRNA34B, miRNA34C, PMAIP1, POU4F2, PPP1R13B, TP73, NOXA, AMID and ASPP1 has also been detected. In the same way, hypermethylation of genes such as CDKN1A, CDKN1C, CDKN2A, POU4F1, which participate in cell cycle control dependent on p53, and hypermethylation of LATS2 and DAPK1, which participate in the regulation of this same pathway.4,20,59,71,75

The WNT/β-catenin pathway has been involved amply in diverse types of cancer.76 Activation of this pathway regulates proliferation and cell differentiation, among other processes.4 In ALL, an alteration regarding methylation of genes associated with the WNT, such as WNT5A, RSPO1 and APC has been reported.36,72,77 Additionally, aberrant methylation of sFRP2, sFRP4, sFRP5, WIF1, DKK3, sFRP1, PTPRO, FZ10 and DKK2 genes, as well as in β-catenin inhibitor, cadherins (CDH1, CDH11, CD13), and genes of the SOX family (SOX2, 3, 8, 9, 11, 14, 21) has been reported in patients with ALL-B with relapse.54

Other signaling pathways affected in ALL by alterations in DNA methylation include the ephrin receptors (EPH), which are tyrosine-kinase receptors that activate and regulate diverse biological processes. In ALL, it has been suggested that EPH genes can act as tumor suppressors. Hypermethylation in receptors and ligands, including EPHA2, EPHA4, EPHA5, EPHA6, EPHA7, EPHA10, EPHB1, EPHB2, EPHB3, EPHB4, EFNA1, EFNA3, EFNA5, and EFNAB1 has been reported.36 Additionally, alterations in methylation of the MAP kinase pathway genes (MAP2K1, BRAF, ERK2) have been reported in ALL, as well as in the PI3K-AKT pathway.54

4Histones modification in ALL

Histones are proteins involved in DNA organization inside the nucleus. There are five types of histones: H1/H5, H2A, H2B, H3 y H4. Histones H2A, H2B, H3 and H4 form the nucleosomes which pack the DNA, whereas histone H1 is found in the spaces between the nucleosomes; histone H5 is present in specific regions of the DNA. The state of the chromatin depends on post-translational modifications in histones and influences gene transcriptional states. Numerous covalent histone modifications are known, such as acetylation, methylation and phosphorylation, which lead to gene repression or activation (Figure 1).4,9 The predominant and most studied modifications are lysine and lysine/arginine methylation.9

The main enzymes in charge of regulating histone modification are the methyltransferases (HMTs), demethylases (HDMs), acetyltransferase (HATs) and deacetylases (HDACs).78 The combination of the activity of these enzymes confers a “histone code”, which regulates the topology of the chromatin, the accessibility of promoters and, therefore, regulates the transcriptional activity and other processes such as replication and DNA repair.4,79 Some histone modifications associated to open chromatin and, therefore, to transcriptional activation, are lysine (K) acetylation in histone H3 (H3K4, H3K14, H3K9, H3K27) and histone H4 (H4K5, H4K20) (Figure 1).

The enzymes in charge of histone acetylation are the HATs. Methylation of lysine catalyzed by HMTs can activate or repress transcription; for example, monomethylation of H3K4, H3K79, H3K35 activates transcription. Some marks of closed chromatin and transcriptional repression include the trimethylation of lysine in H3K9 and H3K27 (Figure 1).9 Together with DNA methylation, histone modifications have been implicated in the etiology and progression of diverse types of cancer.80 Mutations in different histone modifying enzymes that lead to the loss or gain of its function have been found. In particular, acetylation removes positive charges in histones, which decreases the interaction of phosphate groups of the DNA (negative charge) with the N-terminal portion of histones (positive charges). The latter leads to relaxation of the chromatin, which is associated with greater DNA accessibility and, therefore, increased transcriptional activity.81 HDACs enzymes include the HDAC1, HDAC11 and sirtuins.82 Reduction in histone acetylation derived from the overexpression of HDACs enzymes is a common event in diverse tumors, including leukemia.81–84

In pediatric ALL, overexpression of histone deacetylases, such as HDAC1, HDAC2, HDAC3, HDAC6, HDAC7 y HDAC8, has been identified in comparison to non-leukemic cells (Figure 2).84,85 Particularly, overexpression of HDAC2 and HDAC5 in ALL-B has been reported,84,86 whereas in ALL-T a greater expression of HDAC1, HDAC4 and HDAC5 has been observed.84 As for prognosis, overexpression of HDAC1, HDAC2, HDAC3, HDAC4, HDAC7, HDAC9 y HDAC11 genes has been associated with an unfavorable prognosis in pediatric ALL.84,85,87 Furthermore, acetylation of the H4 histone has been proposed as a marker of prognosis at diagnosis and in relapses. In adults with ALL, increased levels of H4 acetylation correlated with an increase in survival.88 Additionally, ETV6-RUNX1 has been reported to recruit HDACs in ALL-B, which induces remodeling of the chromatin and, consequently, blocks the transcription of certain genes normally activated by RUNX1.89

Although not all the signaling pathways and cell processes in which the alteration of HDACs have been identified in pediatric ALL, they are known to be involved in processes such as transcription (HDAC1, HDAC3, HDAC4, HDAC6, HDAC7, HDAC8, HDAC9), cell cycle regulation (HDAC1, HDAC2, HDAC3, HDAC4, HDAC6, HDAC7), apoptosis (HDAC1, HDAC2, HDAC3, HDAC6, HDAC7), and p53 (HDAC2) and Notch (HDAC1, HDAC3, HDAC7) signaling pathways (Figure 3).4

In ALL, altered HATs also have been identified. The main HATs include the GNAT MYST, and CBP/p300 families.82 Some of these proteins have been involved in chromosomal rearrangements capable of leading to leukemic transformation through alteration of histone acetylation and, consequently, in gene expression.90 Some HATs that have been identified to be overexpressed in ALL-B include KAT7, KAT2A, CREBBP (CPB or KAT3A) and KAT6B.86 This modification has been associated with cell processes such as transcription regulation, proliferation and apoptosis.86 Additionally, it has been observed that KAT2A acetylates and stabilizes the oncoprotein TCF3-PBX1 in ALL cells.91 It has also been found that mutations in the CREBBP gene, which lead to a transcriptional deregulation of its target genes, are associated to pediatric ALL-B relapse.92,93 It has been determined that the overexpression of CREBBP can confer a bad prognosis, including those ALL-B subtypes with standard risk, such as hyperdiploidy.93

The most studied HMT in ALL is MLL, which methylates lysine 4 of the H3 histone (H3K4), which is a mark of open chromatin and, therefore, favors transcription.4 Additionally, MLL regulates transcription through recruitment of HAT protein such as CBP and MOF. HOXA genes are mainly part of MLL multiple targets. The MLL forms rearrangements with over 85 different genes, which lead to the formation of chimeric proteins. Although they lose the active methyltransferase H3K4 domain, they retain the capacity of binding to chromatin.94,95 MLL oncoproteins lead to the aberrant activation of their target genes through diverse mechanisms. For example, the chimeric protein MLL-AF4 alters the regulation of its target genes by recruiting epigenetic regulation complexes such as SEC and DOT1 (H3K79 methyltransferase). This dysregulation leads to leukemic transformation since MEIS1, RUNX1, FLT3, MYC, BCL2, y PROM1 genes are among the targets of MLL-AF4.96 Moreover, MLL has been found fused to HAT proteins, such as CREBBP and EP300, leading to the overexpression of their target genes.96,97

5Alteration in miRNAs

MicroRNAs (miRNAs) constitute another type of epigenetic regulation. They are small RNAs of 18-25 nucleotides which mainly inhibit the translation of their target mRNAs through interaction with the 3′UTR in mammals. miRNAs derive from the processing of precursors by the RNAse III-Drosha-Dicer protein complex.98 Gene expression regulation by miRNAs is a common biological process; more than 60% of mRNAs can be regulated by miRNAs.99 miRNAs are implicated in critical biological processes, including growth and cell development, metabolism, proliferation, differentiation, and apoptosis.17 In cancer, it has been observed that miRNAs participate acting as oncogenes (oncomiRNAs), tumor suppressors, and metastasis suppressors or activators.17 Additionally, miRNAs can be regulated by methylation of the DNA and histone modification (Figure 2).100

Multiple biological processes are regulated by miRNAs, including normal and malignant hematopoiesis.101,102 Alterations in miRNA expression have been identified in ALL-B and T in both children and adults.17 miRNA expression profiles have been used in the diagnosis, classification, and prognosis of ALL.103 In pediatric ALL, overexpression of miR222, miR339, miR142-3p, miR128a, miR128b, miR34a, miR146, miR142, miR181, as well as decreased expression of miR140, miR143, miR451, miR373, miR100, miR196b has been reported in comparison to their normal cell counterparts (Table 6). Distinctive miRNA signatures between ALL genetic subtypes have also been reported. In the hyperdiploid subtype, overexpression of miR198, miR222, miR223, miRNA511 and miRNA708 has been detected, whereas the ETV6-RUNX1 subtype presents miR99a, miR100, miR125b, miR383 and miR708 overexpression (Table 6).103–124 ALL subtype with MLL rearrangements is characterized by the overexpression of miR196B and low expression of miR128b and miR221.103,104

Table 6.

Target genes of frequently altered miRNAs in ALL.

miRNA  Target genes  Reference 
Overexpression
miR128a  BMI  Luan et al., 2015103 
miR708  CNTFR, NNAT, GNG12  Luan et al., 2015103 
miR99a  FKBP5, IGFIR/mTOR  Luan et al., 2015103 
miR125b  Trp53inpI  Luan et al., 2015103 
miR223  FOXO  Luan et al., 2015103 
miR222  BBC3, BCL2L11, BMF, BNIP3, CDKN1B, CDKN1C, CREBZF, DDIT4, DKK2, ESR1, FOS, FOXO3, ICAM1, KIT, MMP1, MYBL1, PPP2R2A, PTEN, SOD2, TBK1  108 
miR142-3p  GRα, PKA  Lv et al., 2012109 
miR128b  BMI-1, PTEN, E2F5, TGFβRI  Li et al., 2013110 
miR34a  SMAD4, CCND1, MAP2K1, BCL2, NOTCH2  Tang et al., 2015111 
miR146  TRAF6, IRAK1  Taganov et al., 2006112 
miR142  APC  Isobe et al., 2014113 
miR181  PRCKD, CTDSPL, CAMKK1  Su et al., 2015114 
miR198  BIRC5, FANCD2, EGF, HMGA1, GPSM2, H2AFX, POLD1, HOXB13, HDAC4, SPDYA  Huang et al., 2016115 
miR511  TLR4  Tserel et al., 2011116 
miR383  EDN1 & TYRP1  Mansuri et al., 2016117 
Underexpression
miR140  DNMT1  Takata et al., 2013118 
miR143  DNMT3A  Ng et al., 2009119 
miR451  MIF  Tang et al., 2015120 
miR373  TXNIP, RABEP1, TNFAIP1  Yang et al., 2011121, Wang et al., 2015122 
miR100  SMARCA5, HOXA1  Luan et al., 2015103, Chen et al., 2014123 
miR196  HOXB8, HOXC8, HOXD8, HOXA7, HOXB7, ERG, HMGA2, ANXA1, S100A9, SPRR2C, KRTS  Chen et al., 2011124 

Diverse miRNAs implicated in cell proliferation and apoptosis have been associated with prognosis in patients with ALL. Low expression of miR456, miR708, miR210, as well as overexpression of miR100/99 have been associated with chemotherapy resistance and, therefore, with an unfavorable prognosis. Other miRNAs which low expression is associated with a bad prognosis include miR124a and miR152. Additionally, overexpression of miR92a is associated with an unfavorable prognosis.60,65,125–127

6Epigenetic therapy in ALL

The identification of epigenetic alterations in ALL has motivated the search of molecules capable of reverting these alterations. In patients with ALL, two hypomethylating agents with previous approval in the United States for their use in other types of cancer have been tested. In patients with refractory ALL or in relapses, inhibitors of the DNA-methyltransferase, such as 5-azacitidine and decitabine, in combination with other conventional chemotherapeutic drugs have been used. In these studies, evidence of clinical activity without excessive toxicity was observed.128,129 In addition to demethylating agents, histone inhibitory agents, such as vorinostat and panobinostat, have been tested in patients with ALL. In a phase II study, the administration of decitabine in combination with vorinostat was tested before reinduction chemotherapy in children and adults with refractory ALL. In one out of 13 patients, death by toxicity was registered. In 5/8 patients who completed the treatment, an allogeneic hematopoietic stem cell transplantation was performed, of whom three died of transplant related causes and two survived without evidence of the disease.130

Additionally, it has been proposed that through the regulation of certain miRNAs, an improvement of the therapeutic effect of glucocorticoids could be achieved, particularly for those patients that show resistance to agents such as prednisone. It has been demonstrated that miRNA-17 inhibition leads to an increase in the sensitivity of leukemic cells to dexamethasone.131 In those patients with MLL rearrangements insensitive to glucocorticoids treatment, restitution of the expression of miR128b and miR221 has been suggested as therapy. Both miRNAs show low expression in ALL patients with MLL rearrangements. It has been observed that miR128b has MLL, AF4 genes as targets, and even the oncogenic fusions MLL-AF4 and AF4-MLL. Moreover, miR222 is capable of negatively regulating CDKN1B. In a cooperative manner, both miRNAs are capable of potentiating the sensitivity of leukemic cells to glucocorticoids.132 The restoration of the expression of miR143 has also been proposed as therapy for patients with ALL with the MLL rearrangement; in these patients, miR143 is hypermethylated and silenced. This miRNA has been identified as a regulator of the expression of MLL-AF4. miRNA143 restitution induces apoptosis of leukemic cells.133

In patients with BCR-ABL1-positive ALL, restitution of miR203 has been proposed as therapy. This miRNA has ABL1 and BCR-ABL1 as targets. In BCR-ABL1-positive leukemia, miR203 has been found to be silenced by genetic and epigenetic mechanisms. In patients with BCR-ABL1-positive ALL, who show resistance to tyrosine-kinase inhibitor, it has also been suggested that restitution of miR217 could be used as therapy to inhibit DNTM3A.134,135 It has been reported that patients with BCR-ABL1-positive ALL acquire resistance to tyrosine-kinase inhibitor through the downregulation of miR217, which in turn up-regulates DNMT3A.

7Conclusions

Epigenetic regulation through DNA methylation, histone modification, and by miRNAs plays a fundamental role in the development and evolution of ALL and other types of cancer. In ALL, these levels of regulation have been involved in the etiology of the disease, as well as in the diagnosis, classification, and prognosis of patients. The advent of genome wide association studies allows the detection of an ample number of epigenetic modifications characteristic of ALL, which reflect the complexity and heterogeneity of epigenetic regulation in this disease. Research in this field will keep offering information to allow a better understanding of the complex etiology of ALL. Moreover, knowledge of the epigenetics of ALL will contribute to identify therapeutic targets that could be modified with specific treatments, with the advantage that, unlike genetic modifications, epigenetic alterations are reversible.

Conflict of interests

The authors declare no conflict of interests of any nature.

Acknowledgements

Miguel Alemán 2012 Foundation, Federal Funds 2013 Instituto Nacional de Pediatría, Sectorial Fund of Research for Education SEP-CONACyT CB-2012-01/183467. Navarrete-Meneses thanks to the Biological Sciences Postgraduate Program, CONACyT 385279.

References
[1]
C.G. Mullighan.
Molecular genetics of B-precursor acute lymphoblastic leukemia.
J Clin Invest., 122 (2012), pp. 3407-3415
[2]
C.H. Pui, L.L. Robison, A.T. Look.
Acute lymphoblastic leukaemia.
Lancet., 371 (2008), pp. 1030-1043
[3]
S. Tsuzuki, M. Seto, M. Greaves, T. Enver.
Modeling first-hit functions of the t(12;21) TEL-AML1 translocation in mice.
Proc Natl Acad Sci., 101 (2004), pp. 8443-8448
[4]
E. San Jose-Eneriz, X. Agirre, P. Rodríguez-Otero, F. Prosper.
Epigenetic regulation of cell signaling pathways in acute lymphoblastic leukemia.
Epigenomics., 5 (2013), pp. 525-538
[5]
C.H. Pui.
Acute lymphoblastic leukemia: introduction.
[6]
M. Morrow, S. Horton, D. Kioussis, H. Brady, O. Williams.
TEL-AML1 promotes development of specific hematopoietic lineages consistent with preleukemic activity.
Blood., 103 (2004), pp. 3890-3896
[7]
Q.W. Chen, X.Y. Zhu, Y.Y. Li, Z.Q. Meng.
Epigenetic regulation and cancer (review).
Oncol Rep., 31 (2014), pp. 523-532
[8]
S.B. Baylin, P.A. Jones.
A decade of exploring the cancer epigenome—biological and translational implications.
Nat Rev Cancer., 11 (2011), pp. 726-734
[9]
M.J. Burke, T. Bhatla.
Epigenetic modifications in pediatric acute lymphoblastic leukemia.
Front Pediatr., 2 (2014), pp. 42
[10]
M.A. Dawson, T. Kouzarides.
Cancer epigenetics: from mechanism to therapy.
[11]
C.H. Pui, W.E. Evans.
Treatment of acute lymphoblastic leukemia.
N Engl J Med., 354 (2006), pp. 166-178
[12]
A.F. Hernández, P. Menéndez.
Linking pesticide exposure with pediatric leukemia: potential underlying mechanisms.
Int J Mol Sci., 17 (2016), pp. 461
[13]
W.L. Carroll, D. Bhojwani, D.-J. Min, E. Raetz, M. Relling, S. Davies, et al.
Pediatric acute lymphoblastic leukemia.
Hematology Am Soc Hematol Educ Program., 2003 (2003), pp. 102-131
[14]
C.G. Mullighan.
The molecular genetic makeup of acute lymphoblastic leukemia.
Hematology Am Soc Hematol Educ Program., 2012 (2012), pp. 389-396
[15]
D. Bhojwani, J.J. Yang, C.H. Pui.
Biology of childhood acute lymphoblastic leukemia.
Pediatr Clin North Am., 62 (2015), pp. 47-60
[16]
M.F. Greaves, J. Wiemels.
Origins of chromosome translocations in childhood leukaemia.
Nat Rev Cancer., 3 (2003), pp. 639-649
[17]
Z. Chatterton, L. Morenos, R. Saffery, J.M. Craig, D. Ashley, N.C. Wong.
DNA methylation and miRNA expression profiling in childhood B-cell acute lymphoblastic leukemia.
Epigenomics., 2 (2010), pp. 697-708
[18]
J.A. Timms, C.L. Relton, J. Rankin, G. Strathdee, J.A. McKay.
DNA methylation as a potential mediator of environmental risks in the development of childhood acute lymphoblastic leukemia.
Epigenomics., 8 (2016), pp. 519-536
[19]
L. Scourzic, L. Couronné, M.T. Pedersen, V. Della Valle, M. Diop, E. Mylonas, et al.
DNMT3A(R882H) mutant and Tet2 inactivation cooperate in the deregulation of DNA methylation control to induce lymphoid malignancies in mice.
Leukemia., 30 (2016), pp. 1388-1398
[20]
G. Garcia-Manero, H. Yang, S.-Q. Kuang, S. O’Brien, D. Thomas, H. Kantarjian.
Epigenetics of acute lymphocytic leukemia.
Semin Hematol., 46 (2009), pp. 24-32
[21]
H.Y. Fu, D.S. Wu, H.R. Zhou, J.Z. Shen.
CpG island methylator phenotype and its relationship with prognosis in adult acute leukemia patients.
Hematology., 19 (2014), pp. 329-337
[22]
A. Batova, M.B. Diccianni, J.C. Yu, T. Nobori, M.P. Link, J. Pullen, et al.
Frequent and selective methylation of p15 and deletion of both p15 and p16 in T-cell acute lymphoblastic leukemia.
Cancer Res., 57 (1997), pp. 832-836
[23]
I.H. Wong, M.H. Ng, D.P. Huang, J.C. Lee.
Aberrant p15 promoter methylation in adult and childhood acute leukemias of nearly all morphologic subtypes: potential prognostic implications.
Blood., 95 (2000), pp. 1942-1949
[24]
G. Garcia-Manero, J. Daniel, T.L. Smith, S.M. Kornblau, M.-S. Lee, H.M. Kantarjian, et al.
DNA methylation of multiple promoter-associated CpG islands in adult acute lymphocytic leukemia.
Clin Cancer Res., 8 (2002), pp. 2217-2224
[25]
H. Mai, X. Liu, Y. Chen, C. Li, L. Cao, X. Chen, et al.
Hypermethylation of p15 gene associated with an inferior poor long-term outcome in childhood acute lymphoblastic leukemia.
J Cancer Res Clin Oncol., 142 (2016), pp. 497-504
[26]
D. Mirebeau, C. Acquaviva, S. Suciu, R. Bertin, N. Dastugue, A. Robert, et al.
The prognostic significance of CDKN2A. CDKN2B and MTAP inactivation in B-lineage acute lymphoblastic leukemia of childhood. Results of the EORTC studies 58881 and 58951.
Haematologica., 91 (2006), pp. 881-885
[27]
X. Agirre, J.L. Vizmanos, M.J. Calasanz, M. García-Delgado, M.J. Larráyoz, F.J. Novo.
Methylation of CpG dinucleotides and/or CCWGG motifs at the promoter of TP53 correlates with decreased gene expression in a subset of acute lymphoblastic leukemia patients.
Oncogene., 22 (2003), pp. 1070-1072
[28]
J. Roman, J.A. Castillejo, A. Jimenez, R. Bornstein, M.G. Gonzalez, M. del Carmen Rodriguez, et al.
Hypermethylation of the calcitonin gene in acute lymphoblastic leukaemia is associated with unfavourable clinical outcome.
Br J Haematol., 113 (2001), pp. 329-338
[29]
G. Garcia-Manero, S. Jeha, J. Daniel, J. Williamson, M. Albitar, H.M. Kantarjian, et al.
Aberrant DNA methylation in pediatric patients with acute lymphocytic leukemia.
Cancer., 97 (2003), pp. 695-702
[30]
S. Zheng, X. Ma, L. Zhang, L. Gunn, M.T. Smith, J.L. Wiemels, et al.
Hypermethylation of the 5’ CpG island of the FHIT gene is associated with hyperdiploid and translocation-negative subtypes of pediatric leukemia.
Cancer Res., 64 (2004), pp. 2000-2006
[31]
S. Kawano, C.W. Miller, A.F. Gombart, C.R. Bartram, Y. Matsuo, H. Asou, et al.
Loss of p73 gene expression in leukemias/lymphomas due to hypermethylation.
Blood., 94 (1999), pp. 1113-1120
[32]
P.G. Corn, S.J. Kuerbitz, M.M. van Noesel, M. Esteller, N. Compitello, S.B. Baylin, et al.
Transcriptional silencing of the p73 gene in acute lymphoblastic leukemia and Burkitt's lymphoma is associated with 5’ CpG island methylation.
Cancer Res., 59 (1999), pp. 3352-3356
[33]
P.G. Corn, B.D. Smith, E.S. Ruckdeschel, D. Douglas, S.B. Baylin, J.G. Herman.
E-cadherin expression is silenced by 5’ CpG island methylation in acute leukemia.
Clin Cancer Res., 6 (2000), pp. 4243-4248
[34]
M. Nakamura, K. Sugita, T. Inukai, K. Goi, K. Iijima, T. Tezuka, et al.
p16/MTS1/INK4A gene is frequently inactivated by hypermethylation in childhood acute lymphoblastic leukemia with 11q23 translocation.
Leukemia., 13 (1999), pp. 884-890
[35]
J.P. Issa, B.A. Zehnbauer, C.I. Civin, M.I. Collector, S.J. Sharkis, N.E. Davidson, et al.
The estrogen receptor CpG island is methylated in most hematopoietic neoplasms.
Cancer Res., 56 (1996), pp. 973-977
[36]
S.Q. Kuang, H. Bai, Z.H. Fang, G. Lopez, H. Yang, W. Tong, et al.
Aberrant DNA methylation and epigenetic inactivation of Eph receptor tyrosine kinases and ephrin ligands in acute lymphoblastic leukemia.
Blood., 115 (2010), pp. 2412-2419
[37]
J. Roman-Gomez, J.A. Castillejo, A. Jimenez, M.G. Gonzalez, F. Moreno, M. Rodriguez, C. del, et al.
5’ CpG island hypermethylation is associated with transcriptional silencing of the p21(CIP1/WAF1/SDI1) gene and confers poor prognosis in acute lymphoblastic leukemia.
Blood., 99 (2002), pp. 2291-2296
[38]
L. Shen, Y. Kondo, J.-P. Issa, G. Garcia-Manero.
Lack of p21(CIP1) DNA methylation in acute lymphocytic leukemia.
Blood., 100 (2002), pp. 3432-3433
[39]
G. Bahari, M. Hashemi, M. Naderi, M. Taheri.
TET2 promoter DNA methylation and expression in childhood acute lymphoblastic leukemia.
Asian Pac J Cancer Prev., 17 (2016), pp. 3959-3962
[40]
M.I. Gutiérrez, A.K. Siraj, M. Bhargava, U. Ozbek, S. Banavali, M.A. Chaudhary, et al.
Concurrent methylation of multiple genes in childhood ALL: correlation with phenotype and molecular subgroup.
Leukemia., 17 (2003), pp. 1845-1850
[41]
A.A. Canalli, H. Yang, S. Jeha, K. Hoshino, B. Sanchez-Gonzalez, M. Brandt, et al.
Aberrant DNA methylation of a cell cycle regulatory pathway composed of P73. P15 and P57KIP2 is a rare event in children with acute lymphocytic leukemia.
Leuk Res., 29 (2005), pp. 881-885
[42]
M.I. Gutiérrez, A.K. Siraj, M.M. Ibrahim, A. Hussain, K. Bhatia.
Childhood and adult ALL: differences in epigenetic lesions associated with cell cycle genes.
Am J Hematol., 80 (2005), pp. 158-160
[43]
A. Jiménez-Velasco, J. Román-Gómez, X. Agirre, M. Barrios, G. Navarro, I. Vázquez, et al.
Downregulation of the large tumor suppressor 2 (LATS2/KPM) gene is associated with poor prognosis in acute lymphoblastic leukemia.
Leukemia., 19 (2005), pp. 2347-2350
[44]
K. Hoshino, A. Quintás-Cardama, H. Yang, B. Sanchez-Gonzalez, G. Garcia-Manero.
Aberrant DNA methylation of the Src kinase Hck, but not of Lyn, in Philadelphia chromosome negative acute lymphocytic leukemia.
Leukemia., 21 (2007), pp. 906-911
[45]
E. San José-Enériz, X. Agirre, J. Román-Gómez, L. Cordeu, L. Garate, A. Jiménez-Velasco, et al.
Downregulation of DBC1 expression in acute lymphoblastic leukaemia is mediated by aberrant methylation of its promoter.
Br J Haematol., 134 (2006), pp. 137-144
[46]
M. Murai, M. Toyota, A. Satoh, H. Suzuki, K. Akino, H. Mita, et al.
Aberrant DNA methylation associated with silencing BNIP3 gene expression in haematopoietic tumours.
Br J Cancer., 92 (2005), pp. 1165-1172
[47]
J. Nordlund, C.L. Bäcklin, P. Wahlberg, S. Busche, E.C. Berglund, M.-L. Eloranta, et al.
Genome-wide signatures of differential DNA methylation in pediatric acute lymphoblastic leukemia.
Genome Biol., 14 (2013), pp. r105
[48]
A.S. Gabriel, F.M. Lafta, E.C. Schwalbe, S. Nakjang, S.J. Cockell, A. Iliasova, et al.
Epigenetic landscape correlates with genetic subtype but does not predict outcome in childhood acute lymphoblastic leukemia.
Epigenetics., 10 (2015), pp. 717-726
[49]
K.H. Taylor, K.E. Pena-Hernandez, J.W. Davis, G.L. Arthur, D.J. Duff, H. Shi, et al.
Large-scale CpG methylation analysis identifies novel candidate genes and reveals methylation hotspots in acute lymphoblastic leukemia.
Cancer Res., 67 (2007), pp. 2617-2625
[50]
S.Q. Kuang, W.G. Tong, H. Yang, W. Lin, M.K. Lee, Z.H. Fang, et al.
Genome-wide identification of aberrantly methylated promoter associated CpG islands in acute lymphocytic leukemia.
Leukemia., 22 (2008), pp. 1529-1538
[51]
Z. Chatterton, L. Morenos, F. Mechinaud, D.M. Ashley, J.M. Craig, A. Sexton-Oates, et al.
Epigenetic deregulation in pediatric acute lymphoblastic leukemia.
Epigenetics., 9 (2014), pp. 459-467
[52]
M.E. Figueroa, S.-C. Chen, A.K. Andersson, L.A. Phillips, Y. Li, J. Sotzen, et al.
Integrated genetic and epigenetic analysis of childhood acute lymphoblastic leukemia.
J Clin Invest., 123 (2013), pp. 3099-3111
[53]
J. Nordlund, C.L. Bäcklin, V. Zachariadis, L. Cavelier, J. Dahlberg, I. Öfverholm, et al.
DNA methylation-based subtype prediction for pediatric acute lymphoblastic leukemia.
Clin Epigenetics., 7 (2015), pp. 11
[54]
L.E. Hogan, J.A. Meyer, J. Yang, J. Wang, N. Wong, W. Yang, et al.
Integrated genomic analysis of relapsed childhood acute lymphoblastic leukemia reveals therapeutic strategies.
Blood., 118 (2011), pp. 5218-5226
[55]
S.T. Lee, M.O. Muench, M.E. Fomin, J. Xiao, M. Zhou, A. de Smith, et al.
Epigenetic remodeling in B-cell acute lymphoblastic leukemia occurs in two tracks and employs embryonic stem cell-like signatures.
Nucleic Acids Res., 43 (2015), pp. 2590-2602
[56]
V. Torrano, J. Procter, P. Cardus, M. Greaves, A.M. Ford.
ETV6-RUNX1 promotes survival of early B lineage progenitor cells via a dysregulated erythropoietin receptor.
Blood., 118 (2011), pp. 4910-4918
[57]
D. Bhojwani, D. Pei, J.T. Sandlund, S. Jeha, R.C. Ribeiro, J.E. Rubnitz, et al.
ETV6-RUNX1-positive childhood acute lymphoblastic leukemia: improved outcome with contemporary therapy.
Leukemia., 26 (2012), pp. 265-270
[58]
N.L. Ramakers-van Woerden, R. Pieters, A.H. Loonen, I. Hubeek, E. van Drunen, H.B. Beverloo, et al.
TEL/AML1 gene fusion is related to in vitro drug sensitivity for L-asparaginase in childhood acute lymphoblastic leukemia.
Blood., 96 (2000), pp. 1094-1099
[59]
S. Busche, B. Ge, R. Vidal, J.-F. Spinella, V. Saillour, C. Richer, et al.
Integration of high-resolution methylome and transcriptome analyses to dissect epigenomic changes in childhood acute lymphoblastic leukemia.
Cancer Res., 73 (2013), pp. 4323-4336
[60]
D.J.P.M. Stumpel, P. Schneider, E.H.J. van Roon, J.M. Boer, P. de Lorenzo, M.G. Valsecchi, et al.
Specific promoter methylation identifies different subgroups of MLL-rearranged infant acute lymphoblastic leukemia, influences clinical outcome, and provides therapeutic options.
Blood., 114 (2009), pp. 5490-5498
[61]
E. Schafer, R. Irizarry, S. Negi, E. McIntyre, D. Small, M.E. Figueroa, et al.
Promoter hypermethylation in MLL-r infant acute lymphoblastic leukemia: biology and therapeutic targeting.
Blood., 115 (2010), pp. 4798-4809
[62]
T. Dunwell, L. Hesson, T.A. Rauch, L. Wang, R.E. Clark, A. Dallol, et al.
A genome-wide screen identifies frequently methylated genes in haematological and epithelial cancers.
Mol Cancer., 9 (2010), pp. 44
[63]
M. Iravani, R. Dhat, C.M. Price.
Methylation of the multi tumor suppressor gene-2 (MTS2 CDKN1, p15INK4B) in childhood acute lymphoblastic leukemia.
Oncogene., 15 (1997), pp. 2609-2614
[64]
M. Kim, S.-H. Yim, N.-S. Cho, S.-H. Kang, D.-H. Ko, B. Oh, et al.
Homozygous deletion of CDKN2A (p16, p14) and CDKN2B (p15) genes is a poor prognostic factor in adult but not in childhood B-lineage acute lymphoblastic leukemia: a comparative deletion and hypermethylation study.
Cancer Genet Cytogenet., 195 (2009), pp. 59-65
[65]
X. Agirre, A. Vilas-Zornoza, A. Jiménez-Velasco, J.I. Martin-Subero, L. Cordeu, L. Gárate, et al.
Epigenetic silencing of the tumor suppressor microRNA Hsa-miR-124a regulates CDK6 expression and confers a poor prognosis in acute lymphoblastic leukemia.
Cancer Res., 69 (2009), pp. 4443-4453
[66]
P. Rodriguez-Otero, J. Román-Gómez, A. Vilas-Zornoza, E.S. José-Eneriz, V. Martín-Palanco, J. Rifón, et al.
Deregulation of FGFR1 and CDK6 oncogenic pathways in acute lymphoblastic leukaemia harbouring epigenetic modifications of the MIR9 family.
Br J Haematol., 155 (2011), pp. 73-83
[67]
G. Hütter, M. Kaiser, M. Neumann, M. Mossner, D. Nowak, C.D. Baldus, et al.
Epigenetic regulation of PAX5 expression in acute T-cell lymphoblastic leukemia.
Leuk Res., 35 (2011), pp. 614-619
[68]
S.H. Thathia, S. Ferguson, H.E. Gautrey, S.D. van Otterdijk, M. Hili, V. Rand, et al.
Epigenetic inactivation of TWIST2 in acute lymphoblastic leukemia modulates proliferation, cell survival and chemosensitivity.
Haematologica., 97 (2012), pp. 371-378
[69]
T.M. Abd El-Hamid, G.I. Mossallam, M.A. Sherisher.
The clinical implications of methylated p15 and p73 genes in adult acute lymphoblastic leukemia.
J Egypt Natl Cancer Inst., 22 (2010), pp. 175-184
[70]
G. Strathdee, T.L. Holyoake, A. Sim, A. Parker, D.G. Oscier, J.V. Melo, et al.
Inactivation of HOXA genes by hypermethylation in myeloid and lymphoid malignancy is frequent and associated with poor prognosis.
Clin Cancer Res., 13 (2007), pp. 5048-5055
[71]
A. Vilas-Zornoza, X. Agirre, V. Martín-Palanco, J.I. Martín-Subero, E. San José-Eneriz, L. Garate, et al.
Frequent and simultaneous epigenetic inactivation of TP53 pathway genes in acute lymphoblastic leukemia.
[72]
S. Takeuchi, M. Matsushita, M. Zimmermann, T. Ikezoe, N. Komatsu, T. Seriu, et al.
Clinical significance of aberrant DNA methylation in childhood acute lymphoblastic leukemia.
Leuk Res., 35 (2011), pp. 1345-1349
[73]
J. Nordlund, L. Milani, A. Lundmark, G. Lönnerholm, A.-C. Syvänen.
DNA methylation analysis of bone marrow cells at diagnosis of acute lymphoblastic leukemia and at remission.
[74]
B.J. Aubrey, A. Strasser, G.L. Kelly.
Tumor-suppressor functions of the TP53 pathway.
Cold Spring Harb Perspect Med., 6 (2016), pp. a026062
[75]
K. Bodoor, Y. Haddad, A. Alkhateeb, A. Al-Abbadi, M. Dowairi, A. Magableh, et al.
DNA hypermethylation of cell cycle (p15 and p16) and apoptotic (p14, p53, DAPK and TMS1) genes in peripheral blood of leukemia patients.
Asian Pac J Cancer Prev., 15 (2014), pp. 75-84
[76]
H. Clevers, R. Nusse.
Wnt/β-catenin signaling and disease.
Cell., 149 (2012), pp. 1192-1205
[77]
Ö. Hatırnaz Ng, S. Fırtına, İ. Can, Z. Karakaş, L. Ağaoğlu, Ö. Doğru, et al.
A possible role for WNT5A hypermethylation in pediatric acute lymphoblastic leukemia.
Turk J Hematol., 32 (2015), pp. 127-135
[78]
S. Rea, F. Eisenhaber, D. O’Carroll, B.D. Strahl, Z.W. Sun, M. Schmid, et al.
Regulation of chromatin structure by site-specific histone H3 methyltransferases.
Nature., 406 (2000), pp. 593-599
[79]
N. Patani, W.G. Jiang, R.F. Newbold, K. Mokbel.
Histone-modifier gene expression profiles are associated with pathological and clinical outcomes in human breast cancer.
Anticancer Res., 31 (2011), pp. 4115-4125
[80]
H. Santos-Rosa, C. Caldas.
Chromatin modifier enzymes, the histone code and cancer.
Eur J Cancer., 41 (2005), pp. 2381-2402
[81]
K. Struhl.
Histone acetylation and transcriptional regulatory mechanisms.
Genes Dev., 12 (1998), pp. 599-606
[82]
C. Zhang, J.F. Zhong, A. Stucky, X.-L. Chen, M.F. Press, X. Zhang.
Histone acetylation: novel target for the treatment of acute lymphoblastic leukemia.
Clin Epigenetics., 7 (2015), pp. 117
[83]
A.A. Lane, B.A. Chabner.
Histone deacetylase inhibitors in cancer therapy.
J Clin Oncol., 27 (2009), pp. 5459-5468
[84]
D.A. Moreno, C.A. Scrideli, M.A.A. Cortez, R. de Paula Queiroz, E.T. Valera, V. da Silva Silveira, et al.
Differential expression of HDAC3. HDAC7 and HDAC9 is associated with prognosis and survival in childhood acute lymphoblastic leukaemia.
Br J Haematol., 150 (2010), pp. 665-673
[85]
B. Gruhn, T. Naumann, D. Gruner, M. Walther, S. Wittig, S. Becker, et al.
The expression of histone deacetylase 4 is associated with prednisone poor-response in childhood acute lymphoblastic leukemia.
Leuk Res., 37 (2013), pp. 1200-1207
[86]
Y.-F. Tao, L. Pang, X.-J. Du, L.-C. Sun, S.-Y. Hu, J. Lu, et al.
Differential mRNA expression levels of human histone-modifying enzymes in normal karyotype B cell pediatric acute lymphoblastic leukemia.
Int J Mol Sci., 14 (2013), pp. 3376-3394
[87]
J. Sonnemann, B. Gruhn, S. Wittig, S. Becker, J.F. Beck.
Increased activity of histone deacetylases in childhood acute lymphoblastic leukaemia and acute myeloid leukaemia: support for histone deacetylase inhibitors as antileukaemic agents.
Br J Haematol., 158 (2012), pp. 664-666
[88]
A.S. Advani, S. Gibson, E. Douglas, J. Diacovo, P. Elson, M. Kalaycio, et al.
Histone H4 acetylation by immunohistochemistry and prognosis in relapsed acute lymphocytic leukaemia (ALL).
Br J Haematol., 153 (2011), pp. 504-507
[89]
J. Starkova, J. Madzo, G. Cario, T. Kalina, A. Ford, M. Zaliova, et al.
The identification of (ETV6)/RUNX1-regulated genes in lymphopoiesis using histone deacetylase inhibitors in ETV6/RUNX1-positive lymphoid leukemic cells.
Clin Cancer Res., 13 (2007), pp. 1726-1735
[90]
L. Di Croce.
Chromatin modifying activity of leukaemia associated fusion proteins.
Hum Mol Genet., 14 (2005), pp. R77-R84
[91]
T. Holmlund, M.J. Lindberg, D. Grander, A.E. Wallberg.
GCN5 acetylates and regulates the stability of the oncoprotein E2A-PBX1 in acute lymphoblastic leukemia.
Leukemia., 27 (2013), pp. 578-585
[92]
C.G. Mullighan, J. Zhang, L.H. Kasper, S. Lerach, D. Payne-Turner, L.A. Phillips, et al.
CREBBP mutations in relapsed acute lymphoblastic leukaemia.
Nature., 471 (2011), pp. 235-239
[93]
A. Inthal, P. Zeitlhofer, M. Zeginigg, M. Morak, R. Grausenburger, E. Fronkova, et al.
CREBBP HAT domain mutations prevail in relapse cases of high hyperdiploid childhood acute lymphoblastic leukemia.
Leukemia., 26 (2012), pp. 1797-1803
[94]
C. Meyer, J. Hofmann, T. Burmeister, D. Gröger, T.S. Park, M. Emerenciano, et al.
The MLL recombinome of acute leukemias in 2013.
Leukemia., 27 (2013), pp. 2165-2176
[95]
A.V. Krivtsov, S.A. Armstrong.
MLL translocations, histone modifications and leukaemia stem-cell development.
Nat Rev Cancer., 7 (2007), pp. 823-833
[96]
S. Lin, R.T. Luo, A. Ptasinska, J. Kerry, S.A. Assi, M. Wunderlich, et al.
Instructive role of MLL-fusion proteins revealed by a model of t(4;11) pro-B acute lymphoblastic leukemia.
Cancer Cell., 30 (2016), pp. 737-749
[97]
R.K. Slany.
The molecular biology of mixed lineage leukemia.
Haematologica., 94 (2009), pp. 984-993
[98]
V. Ambros.
The functions of animal microRNAs.
Nature., 431 (2004), pp. 350-355
[99]
R.C. Friedman, K.K.-H. Farh, C.B. Burge, D.P. Bartel.
Most mammalian mRNAs are conserved targets of microRNAs.
Genome Res., 19 (2009), pp. 92-105
[100]
B. Weber, C. Stresemann, B. Brueckner, F. Lyko.
Methylation of human microRNA genes in normal and neoplastic cells.
Cell Cycle., 6 (2007), pp. 1001-1005
[101]
C.-Z. Chen, L. Li, H.F. Lodish, D.P. Bartel.
MicroRNAs modulate hematopoietic lineage differentiation.
Science., 303 (2004), pp. 83-86
[102]
J. Cheng, S. Guo, S. Chen, S.J. Mastriano, C. Liu, A.C. D’Alessio, et al.
An extensive network of TET2-targeting microRNAs regulates malignant hematopoiesis.
Cell Rep., 5 (2013), pp. 471-481
[103]
C. Luan, Z. Yang, B. Chen.
The functional role of microRNA in acute lymphoblastic leukemia: relevance for diagnosis, differential diagnosis, prognosis, and therapy.
OncoTargets Ther., 8 (2015), pp. 2903-2914
[104]
D. Schotte, J.C.K. Chau, G. Sylvester, G. Liu, C. Chen, V.H.J. van der Velden, et al.
Identification of new microRNA genes and aberrant microRNA profiles in childhood acute lymphoblastic leukemia.
Leukemia., 23 (2009), pp. 313-322
[105]
X. Ju, D. Li, Q. Shi, H. Hou, N. Sun, B. Shen.
Differential microRNA expression in childhood B-cell precursor acute lymphoblastic leukemia.
Pediatr Hematol Oncol., 26 (2009), pp. 1-10
[106]
J.C. de Oliveira, C.A. Scrideli, M.S. Brassesco, A.G. Morales, J.A. Pezuk, R. Queiroz, P. de, et al.
Differential miRNA expression in childhood acute lymphoblastic leukemia and association with clinical and biological features.
Leuk Res., 36 (2012), pp. 293-298
[107]
H. Zhang, X.-Q. Luo, P. Zhang, L.-B. Huang, Y.-S. Zheng, J. Wu, et al.
MicroRNA patterns associated with clinical prognostic parameters and CNS relapse prediction in pediatric acute leukemia.
[108]
Human miR-221 & miR-222 Targets. Available at: http://www.sabiosciences.com/rt_pcr_product/HTML/PAHS-6007Z.html.
[109]
M. Lv, X. Zhang, H. Jia, D. Li, B. Zhang, H. Zhang, et al.
An oncogenic role of miR-142-3p in human T-cell acute lymphoblastic leukemia (T-ALL) by targeting glucocorticoid receptor-α and cAMP/PKA pathways.
Leukemia., 26 (2012), pp. 769-777
[110]
M. Li, W. Fu, L. Wo, X. Shu, F. Liu, C. Li.
miR-128 and its target genes in tumorigenesis and metastasis.
Exp Cell Res., 319 (2013), pp. 3059-3064
[111]
T. Tang, R. Su, B. Wang, Y. Zhang.
An integrated approach of predicted miR-34a targets identifies a signature for gastric cancer.
Oncol Lett., 10 (2015), pp. 653-660
[112]
K.D. Taganov, M.P. Boldin, K.-J. Chang, D. Baltimore.
NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses.
Proc Natl Acad Sci U S A., 103 (2006), pp. 12481-12486
[113]
T. Isobe, S. Hisamori, D.J. Hogan, M. Zabala, D.G. Hendrickson, P. Dalerba, et al.
miR-142 regulates the tumorigenicity of human breast cancer stem cells through the canonical WNT signaling pathway.
Elife., (2014), pp. 3
Available at: http://elifesciences.org/lookup/doi/10.7554/eLife.01977
[114]
R. Su, H.-S. Lin, X.-H. Zhang, X.-L. Yin, H.-M. Ning, B. Liu, et al.
MiR-181 family: regulators of myeloid differentiation and acute myeloid leukemia as well as potential therapeutic targets.
Oncogene., 34 (2015), pp. 3226-3239
[115]
W.T. Huang, H.L. Wang, H. Yang, F.-H. Ren, Y.-H. Luo, C.-Q. Huang, et al.
Lower expressed miR-198 and its potential targets in hepatocellular carcinoma: a clinicopathological and in silico study.
OncoTargets Ther., 9 (2016), pp. 5163-5180
[116]
L. Tserel, T. Runnel, K. Kisand, M. Pihlap, L. Bakhoff, R. Kolde, et al.
MicroRNA expression profiles of human blood monocyte-derived dendritic cells and macrophages reveal miR-511 as putative positive regulator of Toll-like receptor 4.
J Biol Chem., 286 (2011), pp. 26487-26495
[117]
M.S. Mansuri, M. Singh, R. Begum.
miRNA signatures and transcriptional regulation of their target genes in vitiligo.
J Dermatol Sci., 84 (2016), pp. 50-58
[118]
A. Takata, M. Otsuka, T. Yoshikawa, T. Kishikawa, Y. Hikiba, S. Obi, et al.
MicroRNA-140 acts as a liver tumor suppressor by controlling NF-κB activity by directly targeting DNA methyltransferase 1 (Dnmt1) expression.
Hepatology., 57 (2013), pp. 162-170
[119]
E.K.O. Ng, W.P. Tsang, S.S.M. Ng, H.C. Jin, J. Yu, J.J. Li, et al.
MicroRNA-143 targets DNA methyltransferases 3A in colorectal cancer.
Br J Cancer., 101 (2009), pp. 699-706
[120]
Y. Tang, W. Wan, L. Wang, S. Ji, J. Zhang.
microRNA-451 inhibited cell proliferation, migration and invasion through regulation of MIF in renal cell carcinoma.
Int J Clin Exp Pathol., 8 (2015), pp. 15611-15621
[121]
G.R. Yan, S.H. Xu, Z.L. Tan, L. Liu, Q.Y. He.
Global identification of miR-373-regulated genes in breast cancer by quantitative proteomics.
Proteomics., 11 (2011), pp. 912-920
[122]
L.-Q. Wang, Y. Zhang, H. Yan, K.-J. Liu, S. Zhang.
MicroRNA-373 functions as an oncogene and targets YOD1 gene in cervical cancer.
Biochem Biophys Res Commun., 459 (2015), pp. 515-520
[123]
D. Chen, Y. Sun, Y. Yuan, Z. Han, P. Zhang, J. Zhang, et al.
miR-100 Induces epithelial-mesenchymal transition but suppresses tumorigenesis, migration and invasion.
PLoS Genet., 10 (2014), pp. e1004177
[124]
C. Chen, Y. Zhang, L. Zhang, S.M. Weakley, Q. Yao.
MicroRNA-196: critical roles and clinical applications in development and cancer.
J Cell Mol Med., 15 (2011), pp. 14-23
[125]
J.H. Ohyashiki, T. Umezu, C. Kobayashi, R.S. Hamamura, M. Tanaka, M. Kuroda, et al.
Impact on cell to plasma ratio of miR-92a in patients with acute leukemia: in vivo assessment of cell to plasma ratio of miR-92a.
BMC Res Notes., 3 (2010), pp. 347
[126]
Y. Mei, C. Gao, K. Wang, L. Cui, W. Li, X. Zhao, et al.
Effect of microRNA-210 on prognosis and response to chemotherapeutic drugs in pediatric acute lymphoblastic leukemia.
Cancer Sci., 105 (2014), pp. 463-472
[127]
B.W. Han, D.D. Feng, Z.G. Li, X.Q. Luo, H. Zhang, X.J. Li, et al.
A set of miRNAs that involve in the pathways of drug resistance and leukemic stem-cell differentiation is associated with the risk of relapse and glucocorticoid response in childhood ALL.
Hum Mol Genet., 20 (2011), pp. 4903-4915
[128]
V.I. Avramis, R.A. Mecum, J. Nyce, D.A. Steele, J.S. Holcenberg.
Pharmacodynamics and DNA methylation studies of high-dose 1-b-D-arabinofuranosyl cytosine before and after in vivo 5-azacytidine treatment in pediatric patients with refractory acute lymphocytic leukemia.
Cancer Chemother Pharmacol., 24 (1989), pp. 203-210
[129]
C.B. Benton, D.A. Thomas, H. Yang, F. Ravandi, M. Rytting, S. O’Brien, et al.
Safety and clinical activity of 5-aza-2’-deoxycytidine (decitabine) with or without Hyper-CVAD in relapsed/refractory acute lymphocytic leukaemia.
Br J Haematol., 167 (2014), pp. 356-365
[130]
M.J. Burke, J.K. Lamba, S. Pounds, X. Cao, Y. Ghodke-Puranaik, B.R. Lindgren, et al.
A therapeutic trial of decitabine and vorinostat in combination with chemotherapy for relapsed/refractory acute lymphoblastic leukemia (ALL).
Am J Hematol., 89 (2014), pp. 889-895
[131]
M. Harada, K. Pokrovskaja-Tamm, S. Söderhäll, M. Heyman, D. Grander, M. Corcoran.
Involvement of miR17 pathway in glucocorticoid-induced cell death in pediatric acute lymphoblastic leukemia.
Leuk Lymphoma., 53 (2012), pp. 2041-2050
[132]
A. Kotani, D. Ha, J. Hsieh, P.K. Rao, D. Schotte, M.L. den Boer, et al.
miR-128b is a potent glucocorticoid sensitizer in MLL-AF4 acute lymphocytic leukemia cells and exerts cooperative effects with miR-221.
Blood., 114 (2009), pp. 4169-4178
[133]
L. Dou, D. Zheng, J. Li, Y. Li, L. Gao, L. Wang, et al.
Methylation-mediated repression of microRNA-143 enhances MLL-AF4 oncogene expression.
Oncogene., 31 (2012), pp. 507-517
[134]
M.J. Bueno, I. Pérez de Castro, M. Gómez de Cedrón, J. Santos, G.A. Calin, J.C. Cigudosa, et al.
Genetic and epigenetic silencing of microRNA-203 enhances ABL1 and BCR-ABL1 oncogene expression.
Cancer Cell., 13 (2008), pp. 496-506
[135]
C. Nishioka, T. Ikezoe, J. Yang, A. Nobumoto, M. Tsuda, A. Yokoyama.
Downregulation of miR-217 correlates with resistance of Ph(+) leukemia cells to ABL tyrosine kinase inhibitors.
Cancer Sci., 105 (2014), pp. 297-307

Please cite this article as: Navarrete-Meneses MdP, Pérez-Vera P. Alteraciones epigenéticas en leucemia linfoblástica aguda. Bol Med Hosp Infant Mex. 2017;74:243–264.

Copyright © 2017. Hospital Infantil de México Federico Gómez
Article options
Tools