Elsevier

Experimental Cell Research

Volume 303, Issue 2, 15 February 2005, Pages 343-359
Experimental Cell Research

dlk acts as a negative regulator of Notch1 activation through interactions with specific EGF-like repeats

https://doi.org/10.1016/j.yexcr.2004.10.001Get rights and content

Abstract

The protein dlk, encoded by the Dlk1 gene, belongs to the Notch epidermal growth factor (EGF)-like family of receptors and ligands, which participate in cell fate decisions during development. The molecular mechanisms by which dlk regulates cell differentiation remain unknown. By using the yeast two-hybrid system, we found that dlk interacts with Notch1 in a specific manner. Moreover, by using luciferase as a reporter gene under the control of a CSL/RBP-Jk/CBF-1-dependent promoter in the dlk-negative, Notch1-positive Balb/c 14 cell line, we found that addition of synthetic dlk EGF-like peptides to the culture medium or forced expression of dlk decreases endogenous Notch activity. Furthermore, the expression of the gene Hes-1, a target for Notch1 activation, diminishes in confluent Balb/c14 cells transfected with an expression construct encoding for the extracellular EGF-like region of dlk. The expression of Dlk1 and Notch1 increases in 3T3-L1 cells maintained in a confluent state for several days, which is associated with a concomitant decrease in Hes-1 expression. On the other hand, the decrease of Dlk1 expression in 3T3-L1 cells by antisense cDNA transfection is associated with an increase in Hes-1 expression. These results suggest that dlk functionally interacts in vivo with Notch1, which may lead to the regulation of differentiation processes modulated by Notch1 activation and signaling, including adipogenesis.

Introduction

The mammalian protein dlk is a cell-surface transmembrane protein containing six EGF-like repeats in the extracellular domain, a transmembrane region, and a short intracellular tail [1], [2], [3], [4], [5], [6], [7]. dlk, encoded by the paternally imprinted gene Dlk1, is a member of the epidermal growth factor (EGF)-like family of homeotic proteins, which includes Notch receptors and their ligands [1], [2]. The proteins of this family mediate protein–protein interactions through their EGF-like repeats and regulate cell fate and differentiation decisions during development in many organisms [1], [8], [9].

The level of expression of Dlk1 is critical to modulate the extracellular signals leading to either cell differentiation or proliferation [10], [11], [12]. dlk is known to participate in several differentiation processes, including adipogenesis [5], [10], [13], [14], hematopoiesis [13], [15], [16], [17], [18], and adrenal gland and neuroendocrine cell differentiation [2], [19], [20], [21], [22]. Recent reports involve dlk in peripheral and central nervous system differentiation [23], [24], in growth arrest and increased malignance of undifferentiated tumors [2], [22], [25], [26], and in wound repair [27]. In addition, mice lacking Dlk1 display growth retardation and accelerated adiposity [28], whereas transgenic mice expressing the complete ectodomain of dlk fused to the human Fc antibody region show both decreased adiposity and glucose tolerance [29]. Several spliced mRNA variants, encoding for dlk proteins possessing or not a protease target sequence, have been reported [5], [30], [31]. The yet unknown protease would promote the release of the extracellular dlk region to the medium as a soluble protein that inhibits adipogenesis [5], [10], [19], [29], [32].

Multiple Notch receptors or ligands have been described in many organisms [8], [9], [33], [34], [35], [36], [37], [38], [39], [40], [41]. These include Drosophila Notch, C. elegans Lin12 and GLP-1, and vertebrate Notch1, 2, 3, and 4. The basic structure of Notch proteins comprises 29–36 epidermal growth factor (EGF)-like motives and three copies of a “Lin-12/Notch repeat” (LNR) in the extracellular region, as well as a RAM (RBP-Jk-associated molecule) domain, a set of six cdc10/Ankyrin repeats, a homopolymer repeat of glutamine (OPA) domain, three nuclear localization signal (NLS) sequences, and a proline–glutamate–serine–threonine-rich (PEST) domain in the intracellular region [9].

Notch ligands include Delta and Serrate in Drosophila, LAG-2 and APX-1 in C. elegans, and the Delta homologs, Dll1, 3, and 4, and the Serrate homologs, Jagged1 and 2, in vertebrates [42], [43]. Notch ligands are transmembrane proteins that share two important extracellular features: a DSL domain at the N-terminus, which interacts with specific EGF-like repeats of Notch proteins [38], [40], [44], [45], and a variable number of EGF-like repeats at their extracellular region. Some of these ligands can be processed by the action of specific proteases that release their extracellular regions to the external medium [46], [47], [48], [49], [50].

Posttranslational modifications and interaction with their ligands trigger a cascade of proteolytic events that finally release the Notch intracellular domain (NICD) from the plasma membrane [47], [51], [52], [53], [54], [55], [56], [57], [58], [59]. NICD translocates then to the nucleus, where it interacts with the DNA-binding protein CSL (CBF-1, suppressor of hairless, LAG-1)/RBP-Jk). NICD displaces a corepressor and binds to CSL/RBP-Jk/CBF-1, generating a transcriptional activation complex that activates several genes [60]. Three of these target genes are Hes-1, Hes-5, and NF-kB2, all of which encode for transcription factors that regulate the expression of other genes involved in differentiation [54], [55], [61]. The CSL/RBP-Jk/CBF-1–Hes-1 pathway is only one among other pathways activated by the action of Notch proteins leading to the regulation of cell differentiation, proliferation, or apoptosis [62], [63]. These alternate pathways involve the p53 family of proteins [64], [65], [66] or the activation of the intracellular protein Deltex [67], [68]. Activation of Deltex inhibits proteins upstream E47 [69], a transcription factor activated by the ras/MAPK pathway, known to be involved in the induction of differentiation or proliferation depending upon extracellular stimuli. A cross-talk between Notch1 and ras/MAPK pathways has been also demonstrated by other investigators [70], [71].

The structure and amino acid sequence of dlk EGF-like repeats are closely related to those present in Dll4 and Delta proteins [1]. However, one important difference between Notch1 ligands and dlk is the lack of a DSL domain at the dlk N-terminus. In this report, we present evidence supporting that, despite lacking a DSL domain, mouse dlk interacts with mouse Notch1 in a specific manner. We also present evidence indicating that these interactions are functional in vivo, supported by the observation of a dlk-dependent inhibition of the Notch signaling pathway leading to the activation of the CSL/RBP-Jk/CBF-1 promoters. The inhibitory effect on Notch signaling was observed in the newly developed Notch1-positive, dlk-negative, Balb/c 14 cell line by enforcing dlk expression through transfection or when synthetic dlk EGF-like peptides were added to the culture medium. Moreover, the expression of Hes-1 decreased in confluent Balb/c 14 cells stably transfected with an extracellular Dlk1 cDNA construct, as well as in 3T3-L1 cells, which show a confluence-related increase in Dlk1 and Notch1 expression levels. Furthermore, confluent 3T3-L1 cells with decreased Dlk1 expression, obtained by transfection with an antisense dlk cDNA expression construct, show an increase in Hes-1 expression compared with nontransfected 3T3-L1 cells.

Our results suggest that dlk may function as a molecule involved in the negative regulation of Notch1 signaling during several differentiation processes regulated by Notch1 activity, including adipogenesis, B and T cell differentiation, and hematopoiesis.

Section snippets

Plasmid constructs

pAS2-1 and pACT2 vectors (Clontech, Palo Alto, CA, USA) were used to obtain yeast GAL4-AD-Dlk1 and GAL4-BD-Notch1 fusion plasmids required for yeast two-hybrid experiments (Fig. 1, Table 1). Plasmid DNA or total cDNA from confluent 3T3-L1 cells were used as templates to amplify different fragments of Dlk1 and Notch1 cDNAs by PCR, using native or recombinant Pfu DNA polymerase (Stratagene, La Jolla, CA, USA). Oligonucleotide primers were designed to contain an EcoRI restriction site at the 5′

The tandem EGF-like repeats 10/11 and 12/13 of mouse Notch1 interact with the extracellular region of mouse dlk in the yeast GAL4 two-hybrid system

The levels of expression of both Dlk1 and Notch1 are critical for adipocyte differentiation of 3T3-L1 fibroblasts in response to IGF-I/insulin [10]. Because the proteins encoded by these genes are transmembrane proteins and possess EGF-like repeats in their extracellular regions, we decided to investigate whether dlk, in spite of lacking a DSL domain, could interact with Notch1 through their EGF-like repeats. For this purpose, we performed two-hybrid system assays using different constructs of

Discussion

To date, very little is known about the molecular mechanisms of dlk action. dlk structure and expression pattern suggest that it could function similarly to other members of the EGF-like homeotic protein family, triggering a specific signal through the interaction of its EGF-like repeats with specific domains of other proteins. However, dlk lacks specific functional domains characteristic of receptors or ligands belonging to this family [1], which has increased the difficulty to establishing

Acknowledgments

We would like to thank Dr. Channa Fuchs, Division of Monoclonal Antibodies, CBER (FDA), for providing the mouse antibody against Notch1 tandem EGF-like repeats 11 and 12. We also would like to thank Beatriz Sánchez-Solana and Amparo Ruvira from the Biochemistry and Molecular Biology Branch, Medical School, University of Castilla-La Mancha, Albacete, Spain, for their help in cell culture and RT–PCR analysis. Finally, we also would like to thank Eva María Monsalve and María Angeles Ballesteros

References (109)

  • G.E. Gray et al.

    Human ligands of the Notch receptor

    Am. J. Pathol.

    (1999)
  • K. Shimizu et al.

    Mouse jagged1 physically interacts with notch2 and other notch receptors. Assessment by quantitative methods

    J. Biol. Chem.

    (1999)
  • K. Shimizu et al.

    Functional diversity among Notch1, Notch2, and Notch3 receptors

    Biochem. Biophys. Res. Commun.

    (2002)
  • K. Shimizu et al.

    Physical interaction of Delta1, Jagged1, and Jagged2 with Notch1 and Notch3 receptors

    Biochem. Biophys. Res. Commun.

    (2000)
  • R.G. Fehon et al.

    Molecular interactions between the protein products of the neurogenic loci Notch and Delta, two EGF-homologous genes in Drosophila

    Cell

    (1990)
  • C.E. Bland et al.

    Notch-induced proteolysis and nuclear localization of the Delta ligand

    J. Biol. Chem.

    (2003)
  • T. Ikeuchi et al.

    The Notch ligands, Delta1 and Jagged2, are substrates for presenilin-dependent “gamma-secretase” cleavage

    J. Biol. Chem.

    (2003)
  • M.J. LaVoie et al.

    The Notch ligands, Jagged and Delta, are sequentially processed by alpha-secretase and presenilin/gamma-secretase and release 2 signaling fragments

    J. Biol. Chem.

    (2003)
  • C.M. Blaumueller et al.

    Intracellular cleavage of Notch leads to a heterodimeric receptor on the plasma membrane

    Cell

    (1997)
  • D. Pan et al.

    Kuzbanian controls proteolytic processing of Notch and mediates lateral inhibition during Drosophila and vertebrate neurogenesis

    Cell

    (1997)
  • C. Brou et al.

    A novel proteolytic cleavage involved in Notch signaling: the role of the disintegrin-metalloprotease TACE

    Mol. Cell

    (2000)
  • J.S. Mumm et al.

    A ligand-induced extracellular cleavage regulates gamma-secretase-like proteolytic activation of Notch1

    Mol. Cell

    (2000)
  • J.C. Aster et al.

    Oncogenic form of NOTCH1 lacking either the primary binding site for RBP-Jκ or nuclear localization sequences retain the ability to associate with RBP-Jκ and activate transcription

    J. Biol. Chem.

    (1997)
  • Y. Sasaki et al.

    The p53 family member genes are involved in the Notch signal pathway

    J. Biol. Chem.

    (2002)
  • C. Garcés et al.

    Notch-1 controls the expression of fatty acid-activated transcription factors and is required for adipogenesis

    J. Biol. Chem.

    (1997)
  • S. Fields

    METHODS: a companion to Meth.

    Enzymology

    (1993)
  • I. Rebay et al.

    Specific EGF repeats of Notch mediate interactions with Delta and Serrate: implications for Notch as a multifunctional receptor

    Cell

    (1991)
  • L.W. Ellisen et al.

    TAN-1, the human homolog of the Drosophila notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms

    Cell

    (1991)
  • C.M. Smas et al.

    Pref-1, a protein containing EGF-like repeats, inhibits adipocyte differentiation

    Cell

    (1993)
  • M.L. Deftos et al.

    Notch signaling in T cell development

    Curr. Opin. Immunol.

    (2000)
  • B. Osborne et al.

    Notch and the immune system

    Immunity

    (1999)
  • A. Soukas et al.

    Distinct transcriptional profiles of adipogenesis in vivo and in vitro

    J. Biol. Chem.

    (2001)
  • M. Guo et al.

    Control of daughter cell fates during asymmetric division: interaction of Numb and Notch

    Neuron

    (1996)
  • E.P. Spana et al.

    Numb antagonizes Notch signaling to specify sibling neuron cell fates

    Neuron

    (1996)
  • Y. Wakamatsu et al.

    NUMB localizes in the basal cortex of mitotic avian neuroepithelial cells and modulates neuronal differentiation by binding to NOTCH-1

    Neuron

    (1999)
  • B.M. Jehn et al.

    c-Cbl binding and ubiquitin-dependent lysosomal degradation of membrane-associated Notch1

    J. Biol. Chem.

    (2002)
  • S.S. Blair

    Notch signaling: fringe really is a glycosyltransferase

    Curr. Biol.

    (2000)
  • J. Laborda

    The role of the epidermal growth factor-like protein dlk in cell differentiation

    Histol. Histopathol.

    (2000)
  • C.H. Jensen et al.

    Protein structure of fetal antigen 1 (FA1). A novel circulating human epidermal-growth-factor-like protein expressed in neuroendocrine 16 tumors and its relation to the gene products of dlk and pG2

    Eur. J. Biochem.

    (1994)
  • M. Paulsen et al.

    Comparative sequence analysis of the imprinted Dlk1-Gtl2 locus in three mammalian species reveals highly conserved genomic 25 elements and refines comparison with the Igf2-H19 region

    Genome Res.

    (2001)
  • C.M. Smas et al.

    Molecular mechanisms of adipocyte differentiation and inhibitory action of pref-1

    Crit. Rev. Eukaryotic Gene Expression

    (1997)
  • A.A. Wylie et al.

    Novel imprinted DLK1/GTL2 domain on human chromosome 14 contains motifs that mimic those 15 implicated in IGF2/H19 regulation

    Genome Res.

    (2000)
  • S. Artavanis-Tsakonas et al.

    Notch signaling

    Science

    (1995)
  • S. Artavanis-Tsakonas et al.

    Notch signaling: cell fate control and signal integration in development

    Science

    (1999)
  • M.J. Ruiz-Hidalgo et al.

    dlk modulates mitogen-activated protein kinase signaling to allow or prevent differentiation

    Exp. Cell Res.

    (2002)
  • S.R. Bauer et al.

    Modulated expression of the epidermal growth factor-like homeotic protein dlk influences stromal-cell–pre-B-cell interactions, stromal cell adipogenesis, and pre-B-cell 18 interleukin-7 requirements

    Mol. Cell. Biol.

    (1998)
  • C.M. Boney et al.

    Regulation of preadipocyte factor-1 gene expression during 3T3-L1 cell differentiation

    Endocrinology

    (1996)
  • M. Kaneta et al.

    A role for pref-1 and HES-1 in thymocyte development

    J. Immunol.

    (2000)
  • K.A. Moore et al.

    Hematopoietic activity of a stromal cell transmembrane protein containing epidermal growth factor-like repeat motifs

    Proc. Natl. Acad. Sci. U. S. A.

    (1997)
  • N. Ohno et al.

    dlk inhibits stem cell factor-induced colony formation of murine hematopoietic progenitors: Hes-1–8 independent effect

    Stem Cells

    (2001)
  • Cited by (0)

    This research was supported by an appointment to the Research Participation Program at the Center for Biologics Evaluation and Research, administered by the Oak Ridge Institute for Science and Education through an interagency agreement between the U. S. Department of Energy and the U. S. Food and Drug Administration. The Spanish Ministry of Education and Culture also supported this research with a fellowship of the Program: “Perfeccionamiento de Tecnólogos en el Extranjero.” This research was also supported by funds from both The Spanish Ministry of Education and Culture and The Regional Ministry of Health of Castilla-La Mancha, Spain.

    View full text