• 沒有找到結果。

mutations that targeted genes in key cellular pathways in B-ALL are identified and are

applied to characterize new subtypes or increase the new insights into known ALL

subtypes, such as BCR/ABL1-like ALL (~15%) (9, 10), intrachromosomal amplification

of chromosome 21 (iAMP21, ~2%) (11, 12), IGH- and CRLF2-rearrangement (5-7%)

(13), ERG-deregulated ALL (~7%) (14), PAX5-deletion (3%) /mutation (5-7%) /

rearrangement (2-3%) (5), and IKZF1-deregulated ALL (~15%) (9, 15).

1.3. ETV6/RUNX1-positive B-ALL

1.3.1. ETV6/RUNX1 fusion gene

The t(12;21) (p13;q22) translocation, which results in ETV6/RUNX1 fusion gene,

was first reported by two different group in 1995 (16, 17). It is the most common

chromosomal rearrangement in childhood B-ALL cases but less prevalent in adult

patients (1, 18). The incidence of t(12;21) in B-ALL is approximately 15-25%, and

patients with this translocation usually have excellent outcome (19). This rearrangement

is not able to be detected by conventional cytogenetic analysis but is readily detected by

fluorescent in situ hybridization and molecular techniques, such as RT-PCR and

quantitative RT-PCR (20-22).

The ETV6 gene, which is very large (240 kb) and consists of eight exons coding for

an ETS-like putative transcription factor containing a helix-loop-helix (HLH) and a

14

DNA-binding domain is on human chromosome 12p13 (23). The RUNX1 gene, which

belongs to the runt domain gene family of transcription factors, is on human

chromosome 21q22 and spans 260 kb consisting of 12 exons (24). Both ETV6 and

RUNX1 genes demonstrated critical roles in hematopoiesis in knockout mice studies

(25-27), and they are also frequently targeted by rearrangements and mutations in

leukemia (28, 29).

The breakpoints on ETV6 gene are clustered in a 15 kb region between exon 5 and

6 (23), and the breakpoints on RUNX1 gene may occur either in the ~100 kb intron 1 or

intron 2 (Appendix IIA) (18, 30). Most of all, the ETV6/RUNX1 fusion transcript shows

a joining of exon 5 of ETV6 to the second exon of RUNX1, while the junction occurred

at the third exon of RUNX1 is less frequently seen (Appendix IIB) (30). Wherever the

breakpoints occurred, these all result in fusion of the 5ʹ portion of ETV6 and almost the

entire coding region of RUNX1 gene.

1.3.2. Structure and function of ETV6/RUNX1 fusion protein

The ETV6/RUNX1 fusion protein is composed of the N-terminal

non-DNA-binding region of ETV6 and nearly complete RUNX1 protein, including the

DNA-binding and activation region, which is responsible for the essential function of

the fusion protein (Appendix III) (16, 18, 31). The ETV6 protein acts as a DNA-binding

15

transcriptional repressor, while RUNX1 can be a DNA-binding transcriptional activator

or repressor depending on the promoter specificity or cell context (18). In contrast to

RUNX1, transiently expressed ETV6/RUNX1 fusion protein generally represses the activities of reporter constructs driven by regulatory regions derived from

hematopoietic-specific genes (18). The ETV6/RUNX1 fusion protein acts as an aberrant

transcription factor that can interfere with the normal functions of wild-type ETV6 and

RUNX1 through multiple mechanisms. ETV6/RUNX1 can dimerize with wild-type

ETV6 through the HLH domain and disrupt the activity of ETV6 (31, 32).

ETV6/RUNX1 may also bind to RUNX1 target DNA sequences and recruit

transcriptional corepressors including mSinA, N-coR, and histone deacetylase-3

(HDAC3) via the fusion part of ETV6, resulting in dysregulated RUNX1-dependent

transcription (Appendix IV) (31, 33, 34).

1.3.3. Role of ETV6/RUNX1 in leukemogenesis of B-ALL

Analysis of monozygotic twins with concordant leukemia and retrospective

screening of neonatal blood spots of patients with leukemia indicate that chromosomal

translocations characteristic of pediatric leukemia often arise prenatally (35). However,

in normal cord blood ETV6/RUNX1 is detected at a frequency that is 100-fold greater

than the risk of the corresponding leukemia (36). Mouse models demonstrate that

16

expression of ETV6/RUNX1 in murine bone morrow stem cells impedes B cell

differentiation from the earliest stages of B cell development with a particular marked impact at the transition from pro-B to pre-B cell stages. Although the accumulation of

both multipotent and B-cell progenitors in vivo, ETV6/RUNX1 is insufficient to induce

leukemia by itself (37, 38). Collectively, these data suggests that ETV6/RUNX1 is a

frequent prenatal first hit in childhood leukemia which can initiate a preleukemic

phenotype remaining covert for up to 15 years but is insufficient for clinical leukemia.

1.4. MicroRNAs

1.4.1. Overview

MicroRNAs (miRNAs) are a group of single-stranded, endogenously initiated

non-coding RNAs which are first discovered in the early 1990s (39). They are

transcribed by RNA polymerase II as part of capped and polyadenylated primary

transcripts (pri-miRNAs) that can be either protein-coding or non-coding. The primary

transcript is cleaved by the Drosha ribonuclease III enzyme to produce an

approximately 70 to 100- nucleotide stem-loop precursor miRNA (pre-miRNA), which

is exported to cytoplasm and is further cleaved by the Dicer ribonuclease to generate the 20-23 nucleotides mature miRNA products from 3ʹ and/or 5ʹ arms (40). When two

mature miRNAs originate from opposite arms of the same pre-miRNA and express

17

similar amounts, they are represented as -3p and -5p (41). In human cells, the mature

miRNAs incorporate into a RNA-induced silencing complex (RISC) and then target

mRNAs for degradation or translational repression via partial or perfect

complementarity to the mRNA 3' untranslated region (3' UTR) through specific seed

sequences (Appendix V) (40, 42). By this way, miRNAs can downregulate gene

expression at the post-transcriptional level. In the recent years, there has been increasing

evidence that miRNAs also bind in the coding region to inhibit translation (43),

implying that miRNAs may flexibly tune the time-scale and magnitude of

post-transcriptional regulation through combination of multiple ways (44). Thus,

miRNAs have the ability to control fundamental cellular functions such as survival,

differentiation, apoptosis, and cell cycle.

1.4.2. MicroRNAs in hematopoiesis and leukemogenesis

A variety of miRNAs have been identified as important regulators of

hematopoiesis (45). For instance, miR-221/222 inhibits the erythropoiesis (46),

miR-223 is essential to modulate myeloid differentiation (47), miR-181a and miR-150

are dynamically regulated during T-cell and B-cell development, respectively, and

premature expression of certain miRNAs in hematopoietic progenitors may impair T-

and B-cell development at the stage transition during maturation (48-50).

18

MiRNA expression in hematological malignancies has also been extensively

studied. Some information about factors that modulate miRNA expression is now

available. Dysregulation of miRNA expression is frequently associated with cytogenetic

abnormalities, and indeed certain abnormalities have direct impacts on aberrant

miRNAs expressions (45). RUNX1/ETO, the most common acute myeloid leukemia–

associated fusion protein resulting from t(8;21), was first reported to directly repress

miR-223 expression by triggering chromatin remodeling and epigenetic silencing,

which in turn blocks the differentiation of myeloid precursor cells (51).

1.4.3. MIR181A1 gene

MIR181A1 gene is located on human chromosome 1q32.1 and is only 62 bp distant

from MIR181B1 gene, they are considered sharing the same primary transcript. There is

only one exon in MIR181A1 gene and expressed a pre-miRNA with 110 bp in length

referred to hsa-mir-181a-1. Hsa-mir-181a-1 will further generate two mature miRNAs

including miR-181a-3p and -5p, which are referred to miR-181a-1 and miR-181a,

respectively (Figure 1A). MIR181A1 belongs to the miR-181 family which consists of

mir-181a/b1, mir-181a/b2 and mir-181c/d, producing four highly similar mature 5p

miRNAs (miR-181a, miR-181b, miR-181c and miR-181d, respectively) with identical

seed sequence and a slight difference in 1~4 bp from three polycistronic transcripts

19

(Figure 1B).

Although both 3p and 5p of hsa-mir-181a can be detected in tissues, previous

reports only demonstrated that miR-181a targets various mRNAs and has physiological

roles and pathological meanings (52-54), while the function of miR-181a-1 remains

unclear. In healthy cells, miR-181a regulates B-cell development, influences T-cell

sensitivity to antigens by modulating T-cell receptor signaling, and is involved in early

steps of hematopoiesis (55). A tumor suppressor activity of miR-181a is reported in

chronic lymphocytic leukemia (CLL) (56, 57), glioma (53), and astrocytoma (54). In

addition, ectopic expression of miR-181a has been shown to sensitizes acute myeloid

leukemia (AML) cell lines to chemotherapy (58), and enhance the effect of radiation

treatment on malignant glioma cells via down-regulation of the Bcl-2 protein (59).

1.4.4. MicroRNAs associated with ETV6/RUNX1

Recent studies have shown that aberrant miRNA expression also plays an

important role in malignant transformation of ETV6/RUNX1 ALL. A highly expressed

miR-125b-2 cluster was found in ETV6/RUNX1 ALL and may provide leukemic cells

with a survival advantage against growth inhibitory signals in a p53-independent

mechanism (60). In addition, ETV6/RUNX1 was shown to regulate the cellular level of

the Survivin protein and apoptosis via suppression of miR-494 and miR-320a

20

expression by direct binding to the promoter regions of their encoding genes (61).