Elsevier

Leukemia Research

Volume 39, Issue 9, September 2015, Pages 976-983
Leukemia Research

Up-regulated A20 promotes proliferation, regulates cell cycle progression and induces chemotherapy resistance of acute lymphoblastic leukemia cells

https://doi.org/10.1016/j.leukres.2015.06.004Get rights and content

Highlights

  • The protein expression of A20 is upregulated in ALL patients and cell lines.

  • A20 depletion decreases proliferation of ALL cells both in vivo and in vitro.

  • A20 depletion induces cell cycle arrest at G0/G1 stage of ALL cells.

  • A20 depletion increases the sensitivity of ALL cells to chemotherapy.

  • The regulation mechanism of A20 was associated with p-ERK and p53 signal pathways.

Abstract

A20, also known as tumor necrosis factor-α (TNFα)-induced protein 3 (TNFAIP3), has been identified as a key regulator of cell survival in many solid tumors. However, little is known about the protein expression level and function of A20 in acute lymphoblastic leukemia (ALL). In this study, we found that A20 is up-regulated in ALL patients and several cell lines. Knockdown of A20 in Jurkat, Nalm-6, and Reh cells resulted in reduced cell proliferation, which was associated with cell cycle arrest. Phospho-ERK (p-ERK) was also down-regulated, while p53 and p21 were up-regulated in A20 knockdown cells. In addition, A20 knockdown induced apoptosis in Jurkat and Reh cells and enhanced the sensitivity of these cell lines to chemotherapeutic drugs. These results indicate that A20 may stimulate cell proliferation by regulating cell cycle progression. A20 inhibited apoptosis in some types of ALL cells, thereby enhancing their resistance to chemotherapy. This effect was abolished through A20 silencing. These findings suggest that A20 may contribute to the pathogenesis of ALL and that it may be used as a new therapeutic target for ALL treatment.

Introduction

Acute lymphoblastic leukemia (ALL) is a hematological malignancy characterized by an accumulation of immature cells in the bone marrow and peripheral blood. It is recognized as the most common type of childhood leukemia [1], [2]. Although great improvements have been made in the treatment of pediatric leukemia, adult ALL remains a high burden [3]. This burden is partly due to uncontrolled cell proliferation and poor response to chemotherapy in adult patients [4]. Leukemic cells abnormally express numerous genes that are associated with proliferation and apoptosis compared to normal hematopoietic cells, and these genes are thought to be the main cause of drug resistance [5]. An examination of abnormally expressed genes in leukemic cells, and of the pathogenic mechanisms associated with these genes, would therefore have great value for the development of targeted therapies.

The zinc-finger protein A20, also known as tumor necrosis factor-α-induced protein 3(TNFAIP3), was first identified as a cytokine-induced gene in human umbilical vein endothelial cells [6]. Many studies have suggested that A20 is an ubiquitin editing protein that regulates inflammation signaling pathways [7] and negatively regulates the nuclear factor kappa B (NF-κB) activation pathway [8]. Some autoimmune diseases, including systemic lupus erythematosus and rheumatoid arthritis, are closely related with A20. Growing evidence also supports a role for A20 in the pathogenesis of cancer [9]. Dysregulation of A20 expression was observed in many types of solid tumor cells; for example, A20 expression is increased in glioblastoma cells [10], [11], hepatocellular carcinoma cells [12], estrogen receptor(ER)-negative or tamoxifen-resistant ER-positive breast cancer cells [13], [14], undifferentiated nasopharyngeal carcinoma cells, and poorly differentiated head and neck cancer cells [15]. These studies suggest an oncogenic role for A20 in these tumors and also suggest that A20 may be a useful new biomarker. However, little is known about the role of A20 in ALL.

In this study, we investigated the expression and biological function of A20 in several types of ALL cells. The results revealed that there was a significantly higher level of A20 protein in cell lines and in bone marrow mononuclear cells (BMMNC) obtained from ALL patients than in cells obtained from healthy donors. In addition, A20 promoted cell proliferation, induced cell cycle arrest, and reduced the sensitivity of three ALL cell lines to daunorubicin via a mechanism involving the ERK, p53 and p21 signal pathways. However, the role of A20 in different ALL cells varied, and the mechanisms by which A20 acted were not completely consistent. This suggests that the effects of A20 are pleotropic and complex in ALL.

Section snippets

Patients and cell lines

Bone marrow samples were obtained from 5 newly diagnosed adult T-ALL or B-ALL patients who were enrolled in the Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, from January 2014 to January 2015. Control bone marrow samples were obtained from 3 healthy hematopoietic stem cell transplantation donors. Diagnosis and leukemia classification were based on 2008 World Health Organization criteria. All subjects provided informed

A20 is expressed at high levels in ALL patients and cell lines

To investigate the function of A20 in ALL, we first analyzed the expression of A20 in newly diagnosed ALL patients and healthy donors. Western blot analysis revealed a significant increase in A20 protein expression in BMMNCs in ALL patients compared to healthy donors (Fig. 1A). In addition, analysis of protein and mRNA levels showed that A20 was highly expressed in several lymphoid cell lines, and particularly in ALL cell lines, compared to normal PBMCs (Fig. 1B and C).

A20 silencing reduces cell proliferation

Following our finding

Discussion

ALL is the most common primary cause of cancer-related death in children [1], [2]. However, the mechanisms leading to uncontrolled cell proliferation in these patients remain elusive. Multiple molecular dysfunctions associated with ALL leukemogenesis may enhance tumor growth and chemotherapy resistance, and these have been attributed to Notch1, MDM2, p53 and microRNAs [17], [18], [19], [20]. Considerable effort has been made to improve our understanding of the pathogenesis of ALL so that we can

Authors’ contributions

Shuying Chen performed most of the experiments, analyzed the data and drafted the manuscript. Haiyan Xing, Shouyun Li, Jing Yu, Huan Li, Shuang Liu, Zheng Tian, Kejing Tang assisted in molecular studies and plasmid construction. Qing Rao and Min Wang contributed to the design of experiments and data interpretation and helped revise the manuscript. Jianxiang Wang contributed to the approval of the final revision of the manuscript.

Acknowledgements

This work was supported by the National Natural Science Foundation of China (Grant No. 81430004, 81370599, 81300380).

References (31)

  • H.F. Tao et al.

    Significance of SODD expression in childhood acute lymphoblastic leukemia and its influence on chemotherapy

    Genet. Mol. Res.

    (2014)
  • S.G. Hymowitz et al.

    A20: from ubiquitin editing to tumour suppression

    Nat. Rev. Cancer

    (2010)
  • Q. Guo et al.

    A20 is overexpressed in glioma cells and may serve as a potential therapeutic target

    Expert Opin. Ther. Targets

    (2009)
  • A.B. Hjelmeland et al.

    Targeting A20 decreases glioma stem cell survival and tumor growth

    PLoS Biol.

    (2010)
  • J.A. Vendrell et al.

    A20/TNFAIP3, a new estrogen-regulated gene that confers tamoxifen resistance in breast cancer cells

    Oncogene

    (2007)
  • Cited by (17)

    • Functional analysis of deubiquitylating enzymes in tumorigenesis and development

      2019, Biochimica et Biophysica Acta - Reviews on Cancer
      Citation Excerpt :

      However, clinical pathological evidence suggests a contradictory result among different types of cancer (Table 9). A20 is overexpressed in both solid and hematological malignancies, such as breast cancer [62], esophageal cancer [318], leukemia [319,320] and glioma [321], where A20 is a negative indicator of patient prognosis. Conversely, A20 is downregulated in a wide spectrum of malignant neoplasms, such as nasopharyngeal cancer, where lower A20 expression correlates to worse survival [322].

    View all citing articles on Scopus
    View full text