Elsevier

Leukemia Research

Volume 31, Issue 6, June 2007, Pages 827-837
Leukemia Research

Derangement of transcription factor profiles during in vitro differentiation of HL60 and NB4 cells

https://doi.org/10.1016/j.leukres.2006.07.019Get rights and content

Abstract

Sequential up- and down-regulation of a handful of critical transcription factors is required for proper neutrophil differentiation. Malfunction of transcription factors may lead to diseases such as acute myeloid leukemia (AML) and specific granule deficiency. In order to understand the molecular background for normal and malignant granulopoiesis, a good model system is required that faithfully mimics the in vivo transcription factor expression profiles. The two human leukemic cell lines HL60 and NB4 have been widely used as model cell lines for these purposes. Differentiation of HL60 and NB4 cells resulted in asynchronous differentiation to morphologically mature neutrophils over a period of 5–7 days. To obtain cell populations of more even maturity, cells at different stages of in vitro differentiation were purified by immunomagnetic isolation. This resulted in three cell populations that could be classified as promyelocytes, myelocytes/metamyelocytes, and mature neutrophils, respectively. Comparison of transcription factor mRNA profiles from these cell populations with those previously seen in normal human bone marrow, demonstrated that although all of the 14 transcription factors described in vivo, could be detected during in vitro differentiation, vast differences in their expression profiles was observed. These data illustrate the limitations of cell lines as models for normal granulopoiesis.

Introduction

Formation of the neutrophil granulocyte takes place in the bone marrow. A number of transcription factors play a critical role during this process and mutations of transcription factor genes may lead to aberrant maturation of neutrophils. Chromosomal translocations involving Runx1 (also called AML1) [1], and mutations or deregulation of C/EBPα [2], [3], [4] is frequently observed in acute myeloid leukemia (AML) patients. Furthermore, mutations of PU.1 have in some cases been associated with AML [5] and disruption of C/EBPɛ has been observed in patients with specific granule deficiency [6].

Gene disruption studies in mice have also identified several transcription factors essential for neutrophil development. Lack of definitive hematopoiesis is observed in Runx1−/− or c-myb−/− fetuses, demonstrating their importance for all hematopoietic lineages, including neutrophils [7], [8], [9]. Mice deficient in C/EBPα completely lack mature neutrophils and eosinophils, whereas all other hematopoietic lineages are present, and function normally [10]. Immature myeloid precursor cells are found in the blood of C/EBPα knock-out mice, demonstrating that C/EBPα is essential for neutrophil differentiation [10]. Conditional knock-outs of C/EBPα have demonstrated that C/EBPα is essential for the transition from the common myeloid progenitor (CMP) to the granulocyte/macrophage restricted progenitor (GMP), but not for GMPs to terminally differentiate to mature neutrophils [11]. C/EBPɛ-deficient mice also fail to develop fully mature neutrophils and eosinophils, only atypical hyposegmented neutrophils were present [12]. Transcripts for azurophil granule proteins were detected in the bone marrow, whereas an almost complete lack of transcripts for specific granule proteins was noted [12], [13], [14]. Mice deficient in PU.1 also generate neutrophils with a block in differentiation [15], [16], [17]. Conditional knock-outs have demonstrated that PU.1 is essential for the generation of CMPs, and for the terminal maturation of GMPs to mature neutrophils [18], [19].

Analysis of knock-out mice is valuable if one wish to demonstrate the requirement of a given transcription factor for myeloid differentiation. However, due to the inherent property of knock-out studies this method only allows one to determine the earliest point of differentiation where the transcription factor in question is essential. Furthermore, as it is becoming evident that not only the presence of a transcription factor, but also its overall concentration [20], [21] – as well as its concentration relative to that of other factor transcription factors [22] – is important for lineage decision and differentiation, model systems to investigate these issues are also required. Cellular systems to study transcription factor regulation during myeloid differentiation in vitro may therefore be of great value provided the models faithfully mimick the in vivo profile of transcription factor expression.

Two human leukemic cell lines, HL60 and NB4, are widely used as models for neutrophil differentiation. The HL60 cell line was established in 1977 from a patient suffering from AML, FAB M2 [23]. The cells grow continuously as myeloblasts and can differentiate to morphologically mature looking neutrophils after addition of all-trans retinoic acid (ATRA) and dimethyl sulfoxide (DMSO) [24]. The differentiation is not complete since specific granules and gelatinase granules are not formed and transcripts for matrix proteins contained in these granules are not detectable [25], [26]. Membrane proteins of these granules are synthesized but are routed to the plasma membrane [27]. Constitutive expression of the specific granule protein NGAL in HL60 cells results in an accumulation of NGAL in the azurophil granules when the cells are at the myeloblast stage. However, when differentiation of HL60 cells is induced, newly synthesized NGAL is routed extracellularly and does not accumulate in granules, reflecting the inability of more mature HL60 cells to synthesize granules and retain granule proteins [25], [27].

The NB4 cell line was established in 1991 from a patient suffering from acute promyelocyte leukemia (APL) having the t(15;17) translocation [28]. Morphologically, the cells are characterized as promyelocytes and can be stimulated to differentiate to neutrophils by ATRA. Like HL60 cells, specific and gelatinase granules are not formed during in vitro differentiation of NB4 cells and mRNA for specific granule matrix proteins cannot be detected [29]. Also like in HL60 cells, membrane proteins of specific and gelatinase granules are synthesized when neutrophil maturation is induced and routed to the plasma membrane [30].

We have previously used a method for separation of neutrophil precursors from human bone marrow into three populations of different maturity. These are myeloblasts (MB) + promyelocytes (PM), myelocytes (MC) + metamyelocytes (MM), and band cells (BC) + segmented neutrophil cells (SC), respectively. By analyzing these populations by Northern and Western blotting, a highly individualized expression of 14 transcription factors important for neutrophil differentiation was demonstrated during in vivo granulopoiesis [31].

The purpose of this study was to correlate the expression-pattern of transcription factor mRNAs in HL60 and NB4 cells during in vitro differentiation, to the in vivo pattern referred to above and to determine whether these two leukemic cell lines may be used as model systems for transcription factor regulation during granulopoiesis.

Section snippets

In vitro differentiation of HL60 and NB4 cells

HL60 cells (CCL-240) were obtained from American Type Culture Collection (ATCC) and NB4 cells were generously provided by Dr. M. Lanotte [28]. The cells were cultured in RPMI 1640 with glutamax-1 (Invitrogen, San Diego, CA, USA) supplemented with 10% fetal calf serum (FCS) (Invitrogen), 100 units/ml penicillin (Invitrogen), and 100 μg/ml streptomycin (Invitrogen). In vitro differentiation of HL60 was induced by addition of 10−6 M ATRA (Sigma–Aldrich, St. Louis, MO, USA) and 1.3% DMSO and of NB4 by

In vitro differentiation of HL60 and NB4 cells and purification by immunomagnetic separation

To investigate the expression of transcription factors during myelopoiesis in vitro, differentiation of HL60 and NB4 cells was induced by addition of 1.3% DMSO + 10−6 M ATRA and 10−6 M ATRA, respectively. Differentiation was not synchronized, as a mixture of cells of different maturity was observed at all times during in vitro differentiation. Even after 6 days of in vitro differentiation some immature cells could be identified although a large number of cells were morphologically mature. To obtain

Discussion

We wished to compare transcription factor mRNA profiles during in vitro differentiation of HL60 and NB4 cells to the pattern observed in vivo. The mRNA profiles previously observed in vivo were very distinct, and steep up- and down-regulations of key factors were observed concomitantly with the transition from one cell population to another [31]. Furthermore, the protein profiles were found to match the mRNA profiles in vivo[31]. When in vitro differentiation of HL60 and NB4 cells was induced,

Acknowledgements

The expert technical assistance of Inge Kobbernagel is greatly appreciated. The authors wish to thank Pia Klausen for fruitful suggestions to the experiments. This work was supported by grants from The Danish Cancer Society, The Danish Medical Research Council, Copenhagen University Hospital (H:S), and The Danish Foundation for Cancer Research.

References (47)

  • M. Lanotte et al.

    NB4, a maturation inducible cell line with t(15;17) marker isolated from a human acute promyelocytic leukemia (M3)

    Blood

    (1991)
  • A. Khanna-Gupta et al.

    NB4 cells show bilineage potential and an aberrant pattern of neutrophil secondary granule protein gene expression

    Blood

    (1994)
  • M.D. Bjerregaard et al.

    The in vivo profile of transcription factors during neutrophil differentiation in human bone marrow

    Blood

    (2003)
  • D.Y. Chih et al.

    Modulation of mRNA expression of a novel human myeloid-selective CCAAT/enhancer binding protein gene (C/EBP epsilon)

    Blood

    (1997)
  • R. Hromas et al.

    A retinoic acid-responsive human zinc finger gene, MZF-1, preferentially expressed in myeloid cells

    J Biol Chem

    (1991)
  • L.M. Scott et al.

    A novel temporal expression pattern of three C/EBP family members in differentiating myelomonocytic cells

    Blood

    (1992)
  • S. Gery et al.

    Retinoic acid regulates C/EBP homologous protein expression (CHOP), which negatively regulates myeloid target genes

    Blood

    (2004)
  • A. Khanna-Gupta et al.

    Chromatin immunoprecipitation (ChIP) studies indicate a role for CCAAT enhancer binding proteins alpha and epsilon (C/EBP alpha and C/EBP epsilon) and CDP/cut in myeloid maturation-induced lactoferrin gene expression

    Blood

    (2003)
  • A.F. Gombart et al.

    Regulation of neutrophil and eosinophil secondary granule gene expression by transcription factors C/EBP epsilon and PU.1

    Blood

    (2003)
  • W. Verbeek et al.

    C/EBPepsilon directly interacts with the DNA binding domain of c-myb and cooperatively activates transcription of myeloid promoters

    Blood

    (1999)
  • A. Khanna-Gupta et al.

    CCAAT displacement protein (CDP/cut) recognizes a silencer element within the lactoferrin gene promoter

    Blood

    (1997)
  • P.M. Lievens et al.

    Repressor activity of CCAAT displacement protein in HL-60 myeloid leukemia cells

    J Biol Chem

    (1995)
  • A. Khanna-Gupta et al.

    Sp1 and C/EBP are necessary to activate the lactoferrin gene promoter during myeloid differentiation

    Blood

    (2000)
  • Cited by (9)

    • MiRNA-130a regulates C/EBP-ε expression during granulopoiesis

      2014, Blood
      Citation Excerpt :

      Furthermore, cells of the pEGP-miR-130a clone showed a more immature morphology with large round nuclei compared with cells of the pEGP-control clone of which some have a donut-shaped nucleus (Figure 4F). FACS analysis showed higher expression of the neutrophil differentiation marker CD11b23 in the pEGP-control clone than in the pEGP-miR-130a clone (Figure 4G), corroborating the more immature phenotype of MPRO cells with high internal miR-130a level. To discriminate between the effect of miR-130a acting through C/EBP-ε and the effect of miR-130a on the expression of other cell cycle proteins, we stably transfected the myeloblast-derived cell line 32Dcl3, which does not express C/EBP-ε,24 with miR-130a.

    • Autoregulatory feedback loop in human hematopoietic cells the c-myb proto-oncogene and microRNA-15a comprise an active

      2009, Blood
      Citation Excerpt :

      With regard to the potential physiologic significance of miR-15a, we found that its expression increased during PMA-induced megakarycytic differentiation of K562 cells (data not shown). A recent study also showed that miR-15a was elevated during ATRA-induced NB4 myeloid differentiation.45,46 It is well documented that c-myb expression is down-regulated during these processes,47 suggesting the possibility that miR-15a may play a biologically relevant role in the inhibition of c-myb expression during hematopoietic cell differentiation.

    • MicroRNA regulation of neutrophil function

      2015, MicroRNAs and Other Non-Coding RNAs in Inflammation
    View all citing articles on Scopus
    View full text