Structure-based mutational analysis of ICAT residues mediating negative regulation of β-catenin co-transcriptional activity

ICAT (Inhibitor of β-CAtenin and TCF) is a small acidic protein that negatively regulates β-catenin co-transcriptional activity by competing with TCF/LEF factors in their binding to β-catenin superhelical core. In melanoma cells, ICAT competes with LEF1 to negatively regulate the M-MITF and NEDD9 target genes. The structure of ICAT consists of two domains: the 3-helix bundle N-terminal domain binds to β-catenin Armadillo (Arm) repeats 10–12 and the C-terminal tail binds to Arm repeats 5–9. To elucidate the structural mechanisms governing ICAT/β-catenin interactions in melanoma cells, three ICAT residues Y15, K19 and V22 in the N-terminal domain, contacting hydrophobic β-catenin residue F660, were mutated and interaction was assessed by immunoprecipitation. Despite the moderate hydrophobicity of the contact, its removal completely abolished the interaction. In the ICAT C-terminal tail consensus sequence, neutralization of the electrostatic interactions between residues D66, E75 and β-catenin residues K435, K312, coupled to deletion of the hydrophobic contact between F71 and β-catenin R386, markedly reduced, but failed to abolish the ICAT-mediated negative regulation of M-MITF and NEDD9 promoters. We conclude that in melanoma cells, anchoring of ICAT N-terminal domain to β-catenin through the hook made by residue F660, trapped in the pincers formed by ICAT residues Y15 and V22, is crucial for stabilizing the ICAT/β-catenin complex. This is a prerequisite for binding of the consensus peptide to Arm repeats 5–9 and competition with LEF1. Differences between ICAT and LEF1 in their affinity for β-catenin may rely on the absence in ICAT of hydrophilic residues between D66 and F71.

[1]  O. Sansom,et al.  E-cadherin can limit the transforming properties of activating β-catenin mutations , 2015, The EMBO journal.

[2]  L. Papon,et al.  β-catenin inhibitor ICAT modulates the invasive motility of melanoma cells. , 2014, Cancer research.

[3]  Moonsup Lee,et al.  PAF and EZH2 induce Wnt/β-catenin signaling hyperactivation. , 2013, Molecular cell.

[4]  F. Rambow,et al.  Beta-catenin inhibits melanocyte migration but induces melanoma metastasis , 2013, Oncogene.

[5]  R. Moon,et al.  WNT signalling pathways as therapeutic targets in cancer , 2012, Nature Reviews Cancer.

[6]  A. Bosserhoff,et al.  WLS inhibits melanoma cell proliferation through the β-catenin signalling pathway and induces spontaneous metastasis , 2012, EMBO molecular medicine.

[7]  Hans Clevers,et al.  Wnt/β-Catenin Signaling and Disease , 2012, Cell.

[8]  Jasmin H. Bavarva,et al.  HEF1, a novel target of Wnt signaling, promotes colonic cell migration and cancer progression , 2011, Oncogene.

[9]  Daniel Bottomly,et al.  Identification of β-catenin binding regions in colon cancer cells using ChIP-Seq , 2010, Nucleic acids research.

[10]  E. Weinberg,et al.  The Terminal Region of β-Catenin Promotes Stability by Shielding the Armadillo Repeats from the Axin-scaffold Destruction Complex* , 2009, The Journal of Biological Chemistry.

[11]  K. Basler,et al.  β-Catenin hits chromatin: regulation of Wnt target gene activation , 2009, Nature Reviews Molecular Cell Biology.

[12]  Jie J. Zheng,et al.  Crystal structure of a full-length beta-catenin. , 2008, Structure.

[13]  R. Dummer,et al.  In vivo switching of human melanoma cells between proliferative and invasive states. , 2008, Cancer research.

[14]  M. Moran,et al.  Large-scale mapping of human protein–protein interactions by mass spectrometry , 2007, Molecular systems biology.

[15]  Jane Goodall,et al.  Mitf regulation of Dia1 controls melanoma proliferation and invasiveness. , 2006, Genes & development.

[16]  D. Fisher,et al.  MITF: master regulator of melanocyte development and melanoma oncogene. , 2006, Trends in molecular medicine.

[17]  L. Chin,et al.  Comparative Oncogenomics Identifies NEDD9 as a Melanoma Metastasis Gene , 2006, Cell.

[18]  W. Weis,et al.  Thermodynamics of beta-catenin-ligand interactions: the roles of the N- and C-terminal tails in modulating binding affinity. , 2006, The Journal of biological chemistry.

[19]  W. Weis,et al.  β-catenin directly displaces Groucho/TLE repressors from Tcf/Lef in Wnt-mediated transcription activation , 2005, Nature Structural &Molecular Biology.

[20]  A. Wittinghofer,et al.  Systematic Peptide Array-based Delineation of the Differential β-Catenin Interaction with Tcf4, E-Cadherin, and Adenomatous Polyposis Coli* , 2005, Journal of Biological Chemistry.

[21]  N. Copeland,et al.  Melanocytes and the microphthalmia transcription factor network. , 2004, Annual review of genetics.

[22]  Nam-Chul Ha,et al.  Mechanism of Phosphorylation-Dependent Binding of APC to β-Catenin and Its Role in β-Catenin Degradation , 2004 .

[23]  T. Akiyama,et al.  Anteriorization of neural fate by inhibitor of beta-catenin and T cell factor (ICAT), a negative regulator of Wnt signaling. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[24]  B. Gumbiner,et al.  Role for ICAT in β-catenin-dependent nuclear signaling and cadherin functions , 2004 .

[25]  Stephen L. Lessnick,et al.  β-Catenin–induced melanoma growth requires the downstream target Microphthalmia-associated transcription factor , 2002, The Journal of Cell Biology.

[26]  W. Weis,et al.  ICAT inhibits beta-catenin binding to Tcf/Lef-family transcription factors and the general coactivator p300 using independent structural modules. , 2002, Molecular cell.

[27]  T. A. Graham,et al.  The crystal structure of the beta-catenin/ICAT complex reveals the inhibitory mechanism of ICAT. , 2002, Molecular cell.

[28]  H. Pehamberger,et al.  The melanocyte-specific isoform of the microphthalmia transcription factor affects the phenotype of human melanoma. , 2002, Cancer research.

[29]  N. Sreerama,et al.  Estimation of protein secondary structure from circular dichroism spectra: comparison of CONTIN, SELCON, and CDSSTR methods with an expanded reference set. , 2000, Analytical biochemistry.

[30]  T. A. Graham,et al.  Crystal Structure of a β-Catenin/Tcf Complex , 2000, Cell.

[31]  H. Varmus,et al.  A comparative evaluation of β-catenin and plakoglobin signaling activity , 2000, Oncogene.

[32]  T. Akiyama,et al.  Inhibition of Wnt signaling by ICAT, a novel β-catenin-interacting protein , 2000, Genes & Development.

[33]  William I. Weis,et al.  Three-Dimensional Structure of the Armadillo Repeat Region of β-Catenin , 1997, Cell.

[34]  K Schulten,et al.  VMD: visual molecular dynamics. , 1996, Journal of molecular graphics.

[35]  P. Argos,et al.  Knowledge‐based protein secondary structure assignment , 1995, Proteins.

[36]  T. Arndt Crystal , 2019, Springer Reference Medizin.

[37]  S. Byers,et al.  Cell-context dependent TCF/LEF expression and function: alternative tales of repression, de-repression and activation potentials. , 2011, Critical reviews in eukaryotic gene expression.

[38]  B. Gumbiner,et al.  Role for ICAT in beta-catenin-dependent nuclear signaling and cadherin functions. , 2004, American journal of physiology. Cell physiology.

[39]  Nam-Chul Ha,et al.  Mechanism of phosphorylation-dependent binding of APC to beta-catenin and its role in beta-catenin degradation. , 2004, Molecular cell.

[40]  R. Shivdasani,et al.  Structure of a human Tcf4-beta-catenin complex. , 2001, Nature structural biology.

[41]  T. Akiyama,et al.  Inhibition of Wnt signaling by ICAT, a novel beta-catenin-interacting protein. , 2000, Genes & development.

[42]  H. Varmus,et al.  A comparative evaluation of beta-catenin and plakoglobin signaling activity. , 2000, Oncogene.

[43]  T. A. Graham,et al.  Crystal structure of a beta-catenin/Tcf complex. , 2000, Cell.