Digital gene expression profiling analysis of childhood acute lymphoblastic leukemia

  • Authors:
    • Ming‑Yi Zhao
    • Yan Yu
    • Min Xie
    • Ming‑Hua Yang
    • Shan Zhu
    • Liang‑Chun Yang
    • Rui Kang
    • Dao‑Lin Tang
    • Ling‑Ling Zhao
    • Li‑Zhi Cao
  • View Affiliations

  • Published online on: April 5, 2016     https://doi.org/10.3892/mmr.2016.5089
  • Pages: 4321-4328
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Acute lymphoblastic leukemia (ALL) is the most commonly diagnosed malignancy in children. It is a heterogeneous disease, and is determined by multiple gene alterations and chromosomal rearrangements. To improve current understanding of the underlying molecular mechanisms of ALL, the present study profiled genome‑wide digital gene expression (DGE) in a population of children with ALL in China. Using second‑generation sequencing technology, the profiling revealed that 2,825 genes were upregulated and 1,952 were downregulated in the ALL group. Based on the DGE profiling data, the present study further investigated seven genes (WT1, RPS26, MSX1, CD70, HOXC4, HOXA5 and HOXC6) using reverse transcription‑quantitative polymerase chain reaction analysis. Gene Ontology analysis suggested that the differentially expressed genes were predominantly involved in immune cell differentiation, metabolic processes and programmed cell death. The results of the present study provided novel insights into the gene expression patterns in children with ALL.

Introduction

Acute lymphoblastic leukemia (ALL) is the most commonly diagnosed malignancy in adolescents and young adults, and represents almost one third of all cases of cancer in children (1,2). ALL is a heterogeneous disease, and multiple subtypes have been identified based on recurrent copy number alterations and structural chromosomal rearrangements (35). Due to genetic variations, the incidence of ALL in children varies across regions, and is determined by ethnicity (1). In China, the incidence of childhood ALL is 4/100,000 children aged <10 years (6). Although the 5-year event-free survival rate of children and adolescents with ALL is ~80%, the remaining 20% of patients relapse, the outcome of which remains poor (79). Therefore, identifying ALL-associated differentially expressed genes is important for improving therapeutic methods and extending patient survival rates in childhood ALL.

Multiple technologies have been used to identify the differentially expressed genes associated with childhood ALL. For example, gene expression microarray analyses of differentially expressed genes in ALL subtypes (10) have identified 80-300 genes as marker genes, which are necessary for discriminating the subtypes. In addition, sequencing-based methods, including serial analysis of gene expression (SAGE), have been used to measure absolute gene expression levels in ALL subtypes (11). However, as hybridization-based methods are subject to technical limitations, certain genes may be overlooked during the hybridization process in gene expression microarray analysis. In addition, the cost of sequencing technology hinders the widespread usage of SAGE for identifying differentially expressed genes (12). Advances in second-generation DNA sequencing technologies has enabled digital gene expression (DGE) profiling to overcome the drawbacks of the microarray analysis hybridization process and allow the detection of differential expression of low-abundance transcripts on a genome-wide scale (13). In the present study, 3′ tag DGE, Gene Ontology (GO) analysis, and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were used to analyze the transcriptional profiles of bone marrow mononuclear cells (BMMCs) from Chinese children with ALL and those without ALL. The results of the analysis revealed numerous gene expression changes attributable to pathogenesis. By investigating changes in the expression of genes, various novel candidate genes required for ALL were identified.

Materials and methods

Patient samples

A total of 10 bone marrow (BM) tissue samples were obtained from children [median age at diagnosis, 8.1 years (range, 3–13.4 years); 5 males and 5 females] newly diagnosed with childhood ALL at Xiangya Hospital (Changsha, China) between May and August 2010. In addition, 10 non-ALL BM samples were obtained from patients who did not have leukemia or other malignancies, but who were undergoing BM aspiration as part of their clinical care at Xiangya Hospital [median follow-up time, 6.75 years (range 4–13.9 years)]. ALL diagnosis was based on the French-American-British classification standards, and the morphology, immunology, cytogenetics and molecular biology classification (14). Complete remission, refractory disease and BM relapse were defined, according to the National Cancer Institute (15). The patient clinical characteristics are listed in Table I. The primary BMMCs from the patients with ALL were analyzed using flow cytometry. The cell-surface antigen staining was performed using a PerCP-conjugated anti-CD45 antibody (cat. no. 304028; BioLegend, Inc., San Diego, CA, USA). For FACS analysis, 5×105 cells were acquired and scored using flow cytometer (Gallios™; Beckman Coulter, Inc., Brea, CA, USA), and the data were analyzed using FlowJo software (version 8.7; Tree Star, Inc., Ashland, OR, USA). The BMMCs were isolated by Ficoll density gradient centrifugation at 671 x g for 20 min at room temperature (Ficoll-Paque was obtained from GE Healthcare Life Sciences; Uppsala, Sweden). The cells at the interface were removed and washed twice with 30 ml sterile phosphate-buffered saline (Well-Biology Co., Ltd., Changsha, China) at 671 x g for 5 min. The isolated cells were counted using the trypan blue (Well-Biology Co., Ltd.,) exclusion method and an inverted phase contrast microscope (IMT-2; Olympus Corporation, Tokyo, Japan). The cells were pelleted and maintained at −80°C until RNA extraction. The RNA was extracted from frozen cell pellets using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA), according to the manufacturer's protocol. In accordance with the Xiangya Hospital Committee on Human Research Review Ethics Committee, written informed consent was obtained from the patients, or from the parents or guardians, as appropriate.

Table I

Clinical characteristics of patients with ALL.

Table I

Clinical characteristics of patients with ALL.

Sample IDImmunophenotypeGenetic subtypeAge at diagnosis (years)WBC count at diagnosis (109 cells/l)Event
Patient 1B-ALLt(12;21)3.0114.2CR1
Patient 2B-ALLt(12;21)13.023.3CR1
Patient 3B-ALLt(12;21)4.26.4CR1
Patient 4B-ALLHeH13.478.3Remission
Patient 5B-ALLHeH3.73.5CR1
Patient 6B-ALLt(9;22)10.233.6CR1
Patient 7T-ALLT-ALL6.0121.5CR1
Patient 8T-ALLT-ALL13.267.8DCR1
Patient 9T-ALLT-ALL2.542.5CR1
Patient 10T-ALLT-ALL10.823.6Remission

[i] ALL diagnosis was established by analyses of morphology, immunophenotype and cytogenetics of leukemic cells. Fluorescence in situ hybridization and/or reverse transcription-polymerase chain reaction analysis were applied to identify t(12;21). ALL, acute lymphoblastic leukemia; CR1, continuous first remission; DCR1, dead in CR1; B-ALL, B-cell lineage ALL; T-ALL, T-cell lineage ALL; t(12;21), translocation between chromosomes 12 and 21 (p13; q22; ETV6/RUNX1); HeH, high hyperdiploidy; t(9;22), translocation between chromosomes 9 and 22) (q11;q34; BCR/ABL1); WBC, white blood cell.

Preparation of sequencing libraries and sequencing

The total RNA sample was digested using DNaseI (M0303S; New England Biolabs, Inc., Ipswich, MA, USA) and purified using oligo-dT beads (Invitrogen Dynabeads mRNA Purification kit; Thermo Fisher Scientific, Inc.), then poly(A)-containing mRNA was fragmented into 130 bp using First-Strand buffer (Thermo Fisher Scientific, Inc.). First-strand cDNA was generated using N6 primer, First Strand Master mix and Super Script II Reverse Transcriptase (Invitrogen; Thermo Fisher Scientific, Inc.) with the following reaction conditions: 25°C for 10 min, 42°C for 40 min and 70°C for 15 min. The Invitrogen Second Strand Master mix (Thermo Fisher Scientific, Inc.) was then added to synthesize the second-strand cDNA (at 16°C for 1 h). The cDNA was purified with Agencourt AMPure XP beads (Beckman Coulter, Inc.), combined with Invitrogen End Repair mix (Thermo Fisher Scientific, Inc.) and incubated at 20°C for 30 min. Invitrogen A-Tailing mix (Thermo Fisher Scientific, Inc.) was added, followed by incubation at 37°C for 30 min. The Adenylate 3′ Ends DNA (Agilent Technologies, Inc., Santa Clara, CA, USA), Illumina adapter (sequence, CATGIAAAAA; Illumina, Inc., San Diego, CA, USA) and ligation mix (Invitrogen; Thermo Fisher Scientific, Inc.) were combined and the ligate reaction was incubated at 20°C for 20 min. Fifteen rounds of PCR amplification were performed with PCR Primer Cocktail and PCR Master mix (both Takara Bio, Inc., Shiga, Japan) to enrich the cDNA fragments. Then the PCR products were purified with Agencourt AMPure XP beads. Sequencing libraries were prepared from 6 µg total RNA using the NlaIII Digital Gene Expression Tag Profiling kit (Illumina, Inc.). The bead-bound cDNA was digested with NlaIII and ligated with the Illumina adaptor (sequence, CATGIAAAAA) containing an MmeI recognition site. The adapter-ligated cDNA was digested with MmeI to release the cDNA from the bead, retaining a 17-bp sequence in the fragment. Following removal of the 3′ fragments via magnetic bead precipitation, a second Illumina adaptor was ligated to the 3′ end of the fragment to form a tagged library. Following 15 cycles of linear PCR amplification, 95-bp fragments were purified by 6% Tris-borate-EDTA polyacrylamide gel electrophoresis (Thermo Fisher Scientific, Inc.). Following denaturation, the single-chain molecules were adhered onto an Illumina Sequencing Chip (Illumina, Inc.). In situ amplification was performed to expand each molecule into a single-molecule cluster sequencing template. Subsequently, four color-labeled nucleotides (Thermo Fisher Scientific, Inc.) were added, and sequencing was performed using the sequencing by synthesis method (16,17). Each tunnel generated millions of raw reads with 35-bp sequencing lengths.

Reverse transcription-quantitative PCR (RT-qPCR) analysis

Total RNA was extracted from the BM using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. The ensuing RT-qPCR was performed using an Access RT-PCR system (ISOGEN; Nippon Gene, Tokyo, Japan), according to the manufacturer's protocol. qPCR was performed using a 25-µl reaction mixture containing 12.5 µl 2X Premix Ex Taq™ (Takara Bio, Inc.), 5 pmol primer and 6.5 µl cDNA, obtained as described above. Amplification was performed on an Applied Biosystem PRISM 7700 system (Thermo Fisher Scientific, Inc.) and the PCR conditions were as follows: 50°C for 2 min, 95°C for 15 min, and 45 cycles at 95°C for 30 sec and 60°C for 1 min, followed by 25°C for 2 min. Quantification was determined by the standard curve and the 2−ΔΔCq method (4). All primer sets were designed, as described previously, synthesized by Biosearch Technologies (Novato, CA, USA) and are presented in Table II.

Table II

Primers used for reverse transcription-quantitative polymerase chain reaction analysis.

Table II

Primers used for reverse transcription-quantitative polymerase chain reaction analysis.

GeneForward primer (5′-3′)Reverse primer (5′-3′)Tm (°C)Product size (bp)
WT1 CTATTCGCAATCAGGGTTA AGGTGGCTCCTAAGTTCAT55312
RPS26 TCCGTGCCTCCAAGATGA ACGCATCGGGCACAGTTA54103
MSX1 CACAAGACGAACCGTAAGCC ACCATATCTTCACCTGCGTCT56154
CD70 TCTCCCGCCTCCCGTAGCAT TGTCCTGCCACCACTACGC56310
HOXC4 CAGACCTCCAGAAATGACG GGGTAGACTATGGGTTGCTT57518
HOXA5 GCAGCACCCACATCAGCA CTTCTGCGGGTCAGGTAA58274
HOXC6 TTCCTACTTCACTAACCCTTCC TGCCCTGCTCAGAACTAAA54323
β-actin GGGTCAGAAGGATTCCTGTG GGTCTCAAACATGATCTGGG54219
Gene annotation

The human transcriptome (Ensembl version 58; http://asia.ensembl.org/) was used as the reference sequence for sequence read alignment and identification. The DGE tags were annotated by mapping the reads to the sequence-flanking NlaIII restriction sites on the coding strands. Alignment and candidate gene identification were performed, as reported previously (18). The total expression profile for each gene was then calculated by summing all the tags mapped to the same gene, including intronic tags.

Statistical analysis

Data were analyzed using GraphPad Prism Software (version 5.0; GraphPad Software, Inc, San Diego, CA, USA). Data are presented as means ± standard deviation or means ± standard error of the mean. Statistical analyses were performed using two-tailed Student's t-tests for the differences between two groups and two-way analysis of variance for the difference between multiple groups. P<0.05 was considered to indicate a statistically significant difference.

Results

mRNA sample preparation and Illumina genome analyses

In the present study, BMMC samples were collected from pediatric patients with ALL (Table I) and from non-ALL individuals. The cellular mRNA was extracted and DGE sequencing libraries were prepared for sequencing in an Illumina Genome Analyzer, which obtained 15,200,000–20,400,000 quality-filtered sequence reads (tags) per sample. Any tags with an abundance of <2 tags per million (TPM), those that mapped to >1 gene, or those that did not match the Ensembl database reference sequence were omitted. Following filtering, 25,000–53,000 unique nucleotide sequence tags were obtained per library, which were mapped to the transcriptome (Fig. 1).

Identification and functional classification of differentially expressed genes

The differentially expressed genes were compared between the patients with childhood ALL and the non-ALL individuals. The selection criteria for putative differentially expressed genes were as follows: i) Average fold change between ALL and non-ALL groups ≥2; ii) single sample t-test false discovery rate <0.1%. In the ALL group, 2,825 genes were upregulated and 1,952 genes were down-regulated (Fig. 2). Of the upregulated genes, 37 that have been associated with the promotion of tumorigenesis, according to Ensembl, were upregulated >10-fold in childhood ALL (Table III).

Table III

Candidate genes upregulated 10-fold in acute lymphoblastic leukemia.

Table III

Candidate genes upregulated 10-fold in acute lymphoblastic leukemia.

Genelog 2 ratioDescriptionFunction
HOXA1112.69349Homeobox A11Endometrial cancer
CCL112.50705Chemokine (C-C motif) ligand 1CC chemokine receptors, airway inflammation
WT112.05494Wilms' tumor 1Wilms' tumor, Leukemia, uterine tumors
TWIST111.80292Twist homolog 1 (Drosophila)Hepatocellular carcinoma
SLITRK610.98513SLIT and NTRK-like family, member 6Leading factor of nerve growth
HLA-DRB410.78627Major histocompatibility complex, class II, DR β4Hepatitis, leukocyte antigen, acute lymphoblastic leukemia
HOXC610.75322Homeobox C6Stem cell differentiation for lymphocytes
ELN10.49685ElastinWilliams syndrome
WIT110.20945Wilms' tumor upstream neighbor 1Wilms' sarcoma downstream factor
SYN110.00000synapsin IDepression neuroblastoma
CD709.911392CD70 moleculeLymphocyte antigen
CTHRC19.850187Collagen triple helix repeat containing 1Gastric cancer, liver cancer, colon cancer
GGT59.850187 γ-glutamyltransferase 5Liver cancer
PROCR9.575539Protein C receptor, endothelialLiver cancer
H199.370687H19, imprinted maternally expressed transcript (non-protein coding)Breast cancer, cervical cancer, choriocarcinoma
HHIP9.326429Hedgehog interacting proteinPancreatic cancer
LIF9.184875Leukemia inhibitory factor (cholinergic differentiation factor)Leukemia inhibitory factor
HOXC49.184875Homeobox C4Stem cell differentiation for lymphocytes
NCR39.134426Natural cytotoxicity triggering receptor 3Multiple myeloma
MSX19.134426Msh homeobox 1Apoptosis
ERBB29.027906v-erb-b2 erythroblastic leukemia viral oncogene homolog 2, neuro/glioblastoma-derived oncogene homolog (avian)Breast, stomach and endometrial cancer, non-small cell lung cancer
LZTS18.851749leucine zipper, putative tumor suppressor 1Primary esophageal cancer
USP27X8.787903Ubiquitin specific peptidase 27, X-linkedUbiquitination
HERV-FRD8.721099HERV-FRD provirus ancestral Env polyproteinRetrovirus
PMS2L18.573647Postmeiotic segregation increased 2-like 1 pseudogeneNasopharyngeal carcinoma
IRX58.495855Iroquois homeobox 5Ovarian cancer
MKRN38.413628Makorin ring finger protein 3Osteocarcinoma
IGFBP68.134426Insulin-like growth factor binding protein 6Breast cancer
TRIM28.027906Tripartite motif-containing 2Cancer cell development
FZD77.912889Frizzled homolog 7 (Drosophila)Wnt signal
NTRK17.912889Neurotrophic tyrosine kinase, receptor, type 1Thyroid cancer
RPS267.912889Ribosomal protein S26Non-Hodgkin's lymphoma
MYCN6.665077v-myc myelocytomatosis viral related oncogene, neuroblastoma derived (avian)Neuroblastoma
SPINK26.427893Serine peptidase inhibitor, Kazal type 2 (acrosin-trypsin inhibitor)Liver cancer
OLIG15.786659Oligodendrocyte transcription factor 1Oligodendrocyte tumor
HOXA75.578747Homeobox A7Epithelial ovarian cancer
NAT145.022015N-acetyltransferase 14 (GCN5-related, putative)Lung cancer
GO analysis

In the present study, GO analysis was performed by mapping each differentially expressed gene into the GO database (http://www.geneontology.org/). The predominant functional group of upregulated genes was associated with the positive regulation of the cellular process, whereas the down-regulated genes were associated with multiple function groups involved in immune cell differentiation, the metabolic process and programmed cell death (Fig. 3).

RT-qPCR analysis of differentially expressed genes

To further evaluate the DGE profiling reliability in the present study, RT-qPCR analysis was performed for a subset of seven genes, which had been determined by the DGE profiling as upregulated >10-fold in the ALL group. The seven genes were expressed at high levels in the ALL samples, and had the same expression profiles as in the initial DGE profiling (Fig. 4). This result confirmed the reliability of the DGE profiling.

Discussion

ALL is a multigenic disease with multiple subtypes; recurrent copy number alterations and structural chromosomal rearrangements characterize the disease. Although the clinical perspective for the ALL subtypes is well established, the underlying molecular mechanisms in the development of ALL development remain to be fully elucidated Thus, char-acterizing ALL-associated differentially expressed genes is important for determining the molecular mechanisms of ALL and for early ALL diagnosis. Several genomic-wide expression profiling approaches have been used to identify the ALL-associated differentially expressed genes (19). The development of second-generation sequencing has enabled the use of DGE profiling for determining genome-wide gene expression profiles in ALL cell samples. For example, Nordlund et al used DGE profiling to characterize the gene expression patterns in different ALL subtypes (20), and found that antisense tags expressed from the non-coding strand were also expressed as a subtype-specific pattern. Additionally, other studies have used microarray-based methods to profile ALL gene expression patterns (10,11,21) However, these previous studies were performed in Western countries, and comparable information in Asian populations is limited. To the best of our knowledge, the present study is the first to use DGE profiling to determine gene expression patterns in childhood ALL in a Chinese population.

The development of second-generation sequencing technology has resulted in DGE profiling possessing several advantages, compared with earlier methods of genome-wide expression analysis (22) For example, it requires smaller RNA samples and costs less than other transcriptome sequencing methods, and it overcomes the limitations of the hybridization process present in microarray-based methods. In addition, computational calculation analysis of DGE data is less challenging. In the present study, DGE profiling revealed that 37 genes were upregulated by >10-fold in the childhood ALL group, compared with the non-ALL group. Notably, these genes are important in tumorigenesis. For example, the wild-type WT1 is expressed in breast cancer, renal cell cancer, ovarian cancer, mesothelioma, lung cancer, melanoma and acute leukemia (23,24). and high levels of WT1 are associated with poor prognosis in ovarian cancer and leukemia (25,26) Other genes, including HOXA11, HHIP, NCR3 and ERBB2 are involved in the tumorigenesis of several types of solid tumor (2731). Thus, in addition to characterizing the expression patterns of ALL-associated genes in the present study, novel candidate genes have been identified, which may be associated with ALL. In addition, GO analysis demonstrated that these genes were predominantly involved in immune cell differentiation, the metabolic process and programmed cell death, suggesting the importance of these signaling pathways in ALL. Future investigations to characterize the roles of these genes in childhood ALL experimentally may further understanding.

In conclusion, DGE profiling was conducted to determine the gene expression profile of childhood ALL in a Chinese population, identifying 2,825 upregulated and 1,952 down-regulated genes. Of these, 37 of the upregulated genes were upregulated by >10-fold, and were found to be important in tumorigenesis. These findings suggested that DGE profiling can provide novel genome-wide information on gene expression in ALL.

Acknowledgments

The present study was supported by grants from The National Natural Sciences Foundation of China (grant nos. 31171328 and 81370648 to Professor Li-Zhi Cao, 81270606 to Dr Yan Yu, 81100359 to Dr Ming-Hua Yang and 81400132 to Dr Shan Zhu) and the Natural Science Foundation of Hunan Province, China (grant no. 2015JJ6118).

References

1 

Biswas S, Chakrabarti S, Chakraborty J, Paul PC, Konar A and Das S: Childhood acute leukemia in West Bengal, India with an emphasis on uncommon clinical features. Asian Pac J Cancer Prev. 10:903–906. 2009.

2 

Burke PW and Douer D: Acute lymphoblastic leukemia in adolescents and young adults. Acta Haematol. 132:264–273. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Awan T, Iqbal Z, Aleem A, Sabir N, Absar M, Rasool M, Tahir AH, Basit S, Khalid AM, Sabar MF, et al: Five most common prognos-tically important fusion oncogenes are detected in the majority of Pakistani pediatric acute lymphoblastic leukemia patients and are strongly associated with disease biology and treatment outcome. Asian Pac J Cancer Prev. 13:5469–5475. 2012. View Article : Google Scholar

4 

Shaikh MS, Adil SN, Shaikh MU and Khurshid M: Frequency of chromosomal abnormalities in Pakistani adults with acute lymphoblastic leukemia. Asian Pac J Cancer Prev. 15:9495–9498. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Zhao M, Yang M, Yang L, Yu Y, Xie M, Zhu S, Kang R, Tang D, Jiang Z, Yuan W, et al: HMGB1 regulates autophagy through increasing transcriptional activities of JNK and ERK in human myeloid leukemia cells. BMB Rep. 44:601–606. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Yeoh AE, Tan D, Li CK, Hori H, Tse E and Pui CH; Asian Oncology Summit 2013: Management of adult and paediatric acute lymphoblastic leukaemia in Asia: Resource-stratified guidelines from the Asian Oncology Summit 2013. Lancet Oncol. 14:e508–e523. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Gaynon PS: Childhood acute lymphoblastic leukaemia and relapse. Br J Haematol. 131:579–587. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Kulkarni KP and Marwaha RK: Outcomes of children with relapsed acute lymphoblastic leukemia in India. Asian Pac J Cancer Prev. 12:1101–1102. 2011.PubMed/NCBI

9 

Tharnprisan P, Khiewyoo J, Sripraya P and Wiangnon S: Relapse-free rate with childhood acute lymphoblastic leukemia treated under the thai national protocol. Asian Pac J Cancer Prev. 14:1127–1130. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Soheila K, Hamid A, Farid Z, Mostafa RT, Nasrin DN, Syyed-Mohammad T and Vahide T: Comparison of univariate and multivariate gene set analysis in acute lymphoblastic leukemia. Asian Pac J Cancer Prev. 14:1629–1633. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Ross ME, Mahfouz R, Onciu M, Liu HC, Zhou X, Song G, Shurtleff SA, Pounds S, Cheng C, Ma J, et al: Gene expression profiling of pediatric acute myelogenous leukemia. Blood. 104:3679–3687. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Unfried C, Burbach G, Korf HW and Von Gall C: Melatonin receptor 1-dependent gene expression in the mouse pars tuberalis as revealed by cDNA microarray analysis and in situ hybridization. J Pineal Res. 48:148–156. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Wang Y, Zhang H, Li H and Miao X: Second-generation sequencing supply an effective way to screen RNAi targets in large scale for potential application in pest insect control. PLoS One. 6:e186442011. View Article : Google Scholar : PubMed/NCBI

14 

Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DA, Gralnick HR and Sultan C: Proposed revised criteria for the classification of acute myeloid leukemia. A report of the French-American-British Cooperative Group. Ann Intern Med. 103:620–625. 1985. View Article : Google Scholar : PubMed/NCBI

15 

Cheson BD, Cassileth PA, Head DR, Schiffer CA, Bennett JM, Bloomfield CD, Brunning R, Gale RP, Grever MR and Keating MJ: Report of the National Cancer Institute-sponsored workshop on definitions of diagnosis and response in acute myeloid leukemia. J Clin Oncol. 8:813–819. 1990.PubMed/NCBI

16 

't Hoen PA, Ariyurek Y, Thygesen HH, Vreugdenhil E, Vossen RH, de Menezes RX, Boer JM, van Ommen GJ and den Dunnen JT: Deep sequencing-based expression analysis shows major advances in robustness, resolution and inter-lab portability over five microarray platforms. Nucleic Acids Res. 36:e1412008. View Article : Google Scholar : PubMed/NCBI

17 

Morrissy AS, Morin RD, Delaney A, Zeng T, McDonald H, Jones S, Zhao Y, Hirst M and Marra MA: Next-generation tag sequencing for cancer gene expression profiling. Genome Res. 19:1825–1835. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Li H, Ruan J and Durbin R: Mapping short DNA sequencing reads and calling variants using mapping quality scores. Genome Res. 18:1851–1858. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Mullighan CG: Genomic profiling of B-progenitor acute lymphoblastic leukemia. Best Pract Res Clin Haematol. 24:489–503. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Nordlund J, Kiialainen A, Karlberg O, Berglund EC, Göransson-Kultima H, Sønderkær M, Nielsen KL, Gustafsson MG, Behrendtz M, Forestier E, et al: Digital gene expression profiling of primary acute lymphoblastic leukemia cells. Leukemia. 26:1218–1227. 2012. View Article : Google Scholar :

21 

Figueroa ME, Chen SC, Andersson AK, Phillips LA, Li Y, Sotzen J, Kundu M, Downing JR, Melnick A and Mullighan CG: Integrated genetic and epigenetic analysis of childhood acute lymphoblastic leukemia. J Clin Invest. 123:3099–3111. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Mullighan CG: Genome sequencing of lymphoid malignancies. Blood. 122:3899–3907. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Iranparast S, Assarehzadegan MA, Heike Y, Hossienzadeh M and Khodadadi A: Wilms' tumor gene (WT1) expression correlates with vascular epithelial growth factor (VEGF) in newly acute leukemia patients undergoing chemotherapy. Asian Pac J Cancer Prev. 15:9217–9223. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Yang L, Han Y, Suarez Saiz F and Minden MD: A tumor suppressor and oncogene: The WT1 story. Leukemia. 21:868–876. 2007.PubMed/NCBI

25 

Bergmann L, Miething C, Maurer U, Brieger J, Karakas T, Weidmann E and Hoelzer D: High levels of Wilms' tumor gene (wt1) mRNA in acute myeloid leukemias are associated with a worse long-term outcome. Blood. 90:1217–1225. 1997.PubMed/NCBI

26 

Netinatsunthorn W, Hanprasertpong J, Dechsukhum C, Leetanaporn R and Geater A: WT1 gene expression as a prognostic marker in advanced serous epithelial ovarian carcinoma: An immunohistochemical study. BMC Cancer. 6:902006. View Article : Google Scholar : PubMed/NCBI

27 

Arteaga CL and Engelman JA: ERBB receptors: From oncogene discovery to basic science to mechanism-based cancer therapeutics. Cancer Cell. 25:282–303. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Hwang JA, Lee BB, Kim Y, Park SE, Heo K, Hong SH, Kim YH, Han J, Shim YM, Lee YS and Kim DH: HOXA11 hypermethylation is associated with progression of non-small cell lung cancer. Oncotarget. 4:2317–2325. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Koch J, Steinle A, Watzl C and Mandelboim O: Activating natural cytotoxicity receptors of natural killer cells in cancer and infection. Trends Immunol. 34:182–191. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Luo LP, Han B, Yu XP, Chen XY, Zhou J, Chen W, Zhu YF, Peng XL, Zou Q and Li SY: Anti-metastasis activity of black rice anthocyanins against breast cancer: Analyses using an ErbB2 positive breast cancer cell line and tumoral xenograft model. Asian Pac J Cancer Prev. 15:6219–6225. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Song Y and Zuo Y: Occurrence of HHIP gene CpG island methylation in gastric cancer. Oncol Lett. 8:2340–2344. 2014.PubMed/NCBI

Related Articles

Journal Cover

May-2016
Volume 13 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao MY, Yu Y, Xie M, Yang MH, Zhu S, Yang LC, Kang R, Tang DL, Zhao LL, Cao LZ, Cao LZ, et al: Digital gene expression profiling analysis of childhood acute lymphoblastic leukemia. Mol Med Rep 13: 4321-4328, 2016
APA
Zhao, M., Yu, Y., Xie, M., Yang, M., Zhu, S., Yang, L. ... Cao, L. (2016). Digital gene expression profiling analysis of childhood acute lymphoblastic leukemia. Molecular Medicine Reports, 13, 4321-4328. https://doi.org/10.3892/mmr.2016.5089
MLA
Zhao, M., Yu, Y., Xie, M., Yang, M., Zhu, S., Yang, L., Kang, R., Tang, D., Zhao, L., Cao, L."Digital gene expression profiling analysis of childhood acute lymphoblastic leukemia". Molecular Medicine Reports 13.5 (2016): 4321-4328.
Chicago
Zhao, M., Yu, Y., Xie, M., Yang, M., Zhu, S., Yang, L., Kang, R., Tang, D., Zhao, L., Cao, L."Digital gene expression profiling analysis of childhood acute lymphoblastic leukemia". Molecular Medicine Reports 13, no. 5 (2016): 4321-4328. https://doi.org/10.3892/mmr.2016.5089