Elsevier

Experimental Cell Research

Volume 313, Issue 14, 15 August 2007, Pages 3141-3152
Experimental Cell Research

Research Article
Notch signaling induces SKP2 expression and promotes reduction of p27Kip1 in T-cell acute lymphoblastic leukemia cell lines

https://doi.org/10.1016/j.yexcr.2007.04.027Get rights and content

Abstract

In T-cell acute lymphoblastic leukemia (T-ALL) NOTCH 1 receptors are frequently mutated. This leads to aberrantly high Notch signaling, but how this translates into deregulated cell cycle control and the transformed cell type is poorly understood. In this report, we analyze downstream responses resulting from the high level of NOTCH 1 signaling in T-ALL. Notch activity, measured immediately downstream of the NOTCH 1 receptor, is high, but expression of the canonical downstream Notch response genes HES 1 and HEY 2 is low both in primary cells from T-ALL patients and in T-ALL cell lines. This suggests that other immediate Notch downstream genes are activated, and we found that Notch signaling controls the levels of expression of the E3 ubiquitin ligase SKP2 and its target protein p27Kip1. We show that in T-ALL cell lines, recruitment of NOTCH 1 intracellular domain (ICD) to the SKP2 promoter was accompanied by high SKP2 and low p27Kip1 protein levels. In contrast, pharmacologically blocking Notch signaling reversed this situation and led to loss of NOTCH 1 ICD occupancy of the SKP2 promoter, decreased SKP2 and increased p27Kip1 expression. T-ALL cells show a rapid G1–S cell cycle transition, while blocked Notch signaling resulted in G0/G1 cell cycle arrest, also observed by transfection of p27Kip1 or, to a smaller extent, a dominant negative SKP2 allele. Collectively, our data suggest that the aberrantly high Notch signaling in T-ALL maintains SKP2 at a high level and reduces p27Kip1, leading to more rapid cell cycle progression.

Introduction

T-cell acute lymphoblastic leukemia (T-ALL) is a malignant disease, originating in thymocytes. T-ALL constitutes a substantial fraction of ALL tumors, both in children and in adults [1]. Current treatment is based on combination chemotherapy, but long-term survival rates are reduced, particularly in older patients, emphasizing the need for improved therapy. The molecular mechanisms underpinning T-ALL are likely to be complex, as a number of genes, including c-MYC, HOX 11, TAL1 and LMO, are involved in chromosomal translocations with the T-cell receptor locus (for review see [1]). Mutations in the Notch signaling pathway have recently emerged as an important genetic component in T-ALL. The involvement of Notch was first observed in rare t(7,9)(q34;q34.3) translocations, which brings an activated form of the NOTCH 1 receptor gene (TAN 1) under control elements from the T-cell receptor gene [2]. More recently, it was shown that more than 50% of all T-ALL patients carry NOTCH1 gain-of-function mutations that, similar to TAN 1, generate an activated form of Notch [3].

The Notch signaling pathway is important for cell fate decisions in many different cell types, including the T-cell lineage [4]. Notch signaling is initiated when the transmembrane Notch receptor interacts with DSL ligands on juxtaposed cells. This leads to two proteolytic processing events in the receptor, first at the extracellular side and subsequently in the plasma membrane. The latter cleavage is executed by the gamma-secretase complex and leads to liberation of the Notch intracellular domain (Notch ICD), which translocates to the nucleus. In the nucleus, Notch ICD binds to the DNA-binding protein CSL, and converts CSL from a repressor to a transcriptional activator [5].

T-ALL mutations lead to aberrantly high Notch signaling and affect two different regions of the NOTCH 1 receptor [3]. One class of mutations falls in the heterodimerization domain (HD), making the receptor more prone to proteolytic processing and thus less dependent on ligand-activation. The other class of mutations occurs in the PEST domain of NOTCH 1, generating truncated versions of the NOTCH 1 ICD, which increases the stability of the normally short-lived intracellular domain [6]. Further mutational analysis of the PEST domain has revealed four serine residues that serve as a negative regulatory sequence, and deletion of these residues makes the Notch ICD more active [7]. The link between deregulated Notch signaling and T-ALL receives further support from mouse models of T-ALL, where Notch 1 mutations occur as secondary hits in mice engineered for deregulation of c-Myc, TAL1, Ikaros and Pbx1 expression [8], [9], [10], [11], [12].

The downstream response of Notch signaling is only partially understood. There is a wealth of data supporting the notion that Hes and Hey genes, which encode bHLH negative transcriptional regulators [13], [14], are immediate downstream genes, but there are situations where Notch signaling exert biological effects without affecting Hes and Hey expression [15]. It is therefore likely that other immediate downstream genes exist that are directly activated by Notch ICD via CSL in specific cellular contexts, and several such genes have recently been identified, e.g. Nrarp, GFAP, GATA2, c-Myc and p21cip1 (see [16] for review).

In T-ALL, our understanding of which downstream genes are activated, and how this affects cell cycle regulation and leads to cellular transformation, is more limited. The finding that HES 1 activation is not always required for Notch-induced growth promotion [17], may suggest that other downstream responses could be relevant for T-ALL. In keeping with this, c-MYC has recently been identified as an important new direct target gene in some T-ALL cell lines [18]. c-MYC was, however, not activated in all T-ALL cell lines [18], suggesting the existence of other immediate Notch downstream effectors.

In this report, we have addressed the downstream aspects of Notch signaling in T-ALL. We find that T-ALL tumors and cell lines have low levels of HES1 and HEY2 expression. Instead, increased Notch signaling results in elevated levels of SKP2, the F-box component in the SCFSkp2 E3 ubiquitin ligase complex [19]. The high level of SKP2 was paralleled by low levels of the CDK inhibitor (CKI) p27Kip1, a target substrate for ubiquitinylation and thus negatively regulated by the SCF–Skp2 complex [20] (for review see [19]). Blocking Notch signaling pharmacologically reversed the situation, and this “Notch off” state was associated with reduced SKP2 and increased p27Kip1 expression and subsequent G1 cell cycle arrest. In conclusion, our data suggest that a NOTCH/SKP2/p27Kip1 axis may contribute to the development of T-ALL.

Section snippets

Cell lines, tumor samples and plasmids

Malignant T-ALL and pre B-ALL cell lines were cultured as described [21]. JM-JURKAT, MOLT4, CEM, peer, RS4 and REH6 cell lines were derived from the ATCC collection. Samples of cryopreserved lymphoblasts were collected from children with the diagnosis of T-ALL and B-precursor-cell phenotype, treated at the Karolinska Hospital, Stockholm, Sweden, and leukemic cells were purified as previously described [22]. Approval was obtained from the Institutional Review Board for these studies. Normal T-

The canonical Notch downstream genes HES1 and HEY2 are expressed at low levels in T-ALL patients and cell lines

We first analyzed whether NOTCH 1 ICD accumulated in T-ALL cell lines, as a reflection of high level Notch signaling. We observed clearly detectable levels of NOTCH 1 ICD in the three T-ALL cell lines JM-JURKAT (JM), MOLT4 and CEM, but not in the B-ALL cell line RS4 (Fig. 1A). The observed bands in the JM and CEM cells were approximately 100 kDa, which corresponds to the expected length of the wild type NOTCH 1 ICD. This is in line with that these cell lines carry missense mutations in the HD

Discussion

In this report we have addressed how the deregulated Notch signaling leads to altered cell cycle progression in T-ALL cells. Our data support the notion that expression of SKP2 and p27Kip1 is under the control of Notch signaling, and that this Notch/SKP2/p27Kip1 axis regulates G1/S cell cycle transition in T-ALL cells. This line of reasoning receives support from several experimental observations. First, the link between Notch and SKP2 is based on two findings: Notch 1 ICD binds to the SKP2

Acknowledgments

We are indebted to Susanne Bergstedt for cell culture work. The financial support from the Swedish Cancer Society, the Swedish Research Council (OS and UL), the EU project EuroStemCell, the Foundation for Strategic Research (CEDB) (UL), the Cancer Society of Stockholm and the Children Cancer Foundation (OS) is gratefully acknowledged.

References (42)

  • M.V. Gustafsson et al.

    Hypoxia requires notch signaling to maintain the undifferentiated cell state

    Dev. Cell

    (2005)
  • J.W. Choi et al.

    Characterization of the transcriptional expression of Notch-1 signaling pathway members, Deltex and HES-1, in developing mouse thymocytes

    Dev. Comp. Immunol.

    (2002)
  • J. Bloom et al.

    Deregulated degradation of the cdk inhibitor p27 and malignant transformation

    Semin. Cancer Biol.

    (2003)
  • S. Yokoi et al.

    A novel target gene, SKP2, within the 5p13 amplicon that is frequently detected in small cell lung cancers

    Am. J. Pathol.

    (2002)
  • K. Nakayama et al.

    Skp2-mediated degradation of p27 regulates progression into mitosis

    Dev. Cell

    (2004)
  • C. Grabher et al.

    Notch 1 activation in the molecular pathogenesis of T-cell acute lymphoblastic leukaemia

    Nat. Rev., Cancer

    (2006)
  • A.P. Weng et al.

    Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia

    Science

    (2004)
  • I. Maillard et al.

    Regulation of lymphoid development, differentiation, and function by the Notch pathway

    Annu. Rev. Immunol.

    (2005)
  • J.C. Aster

    Deregulated NOTCH signaling in acute T-cell lymphoblastic leukemia/lymphoma: new insights, questions, and opportunities

    Int. J. Hematol.

    (2005)
  • M.Y. Chiang et al.

    Identification of a conserved negative regulatory sequence that influences the leukemogenic activity of NOTCH1

    Mol. Cell. Biol.

    (2006)
  • L. Girard et al.

    Frequent provirus insertional mutagenesis of Notch1 in thymomas of MMTVD/myc transgenic mice suggests a collaboration of c-myc and Notch1 for oncogenesis

    Genes Dev.

    (1996)
  • Cited by (97)

    • New insights into TCR β-selection

      2021, Trends in Immunology
      Citation Excerpt :

      Cdkn1b is highly expressed in quiescent ‘pre-β-selection’ DN3 mouse thymocytes, but is downregulated upon initiation of β-selection [41]. Previous studies identified a role for the Skp-Cullin-F-box (SCF) ubiquitin ligase complex, which includes the F-box substrate recognition protein 1 (Fbxl1), also known as S-phase kinase-associated protein 2 (SKP2), in the downregulation of Cdkn1b through ubiquitin-mediated proteasomal degradation of Cdkn1b [19,21,41,47–49]. New data from a mouse model demonstrated additive roles for SCF ubiquitin ligase complexes containing Fbxl1 (or a closely related F-box family member, Fbxl12) in the polyubiquitination and proteasomal degradation of Cdkn1b, by targeting the identical amino acid residue in Cdkn1b (K165) for ubiquitination (Figure 1) [39].

    View all citing articles on Scopus
    View full text