Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 81(1): 57, 2024 Jan 27.
Article in English | MEDLINE | ID: mdl-38279052

ABSTRACT

The Wnt/ß-catenin pathway is critical to maintaining cell fate decisions. Recent study showed that liquid-liquid-phase separation (LLPS) of Axin organized the ß-catenin destruction complex condensates in a normal cellular state. Mutations inactivating the APC gene are found in approximately 80% of all human colorectal cancer (CRC). However, the molecular mechanism of the formation of ß-catenin destruction complex condensates organized by Axin phase separation and how APC mutations impact the condensates are still unclear. Here, we report that the ß-catenin destruction complex, which is constructed by Axin, was assembled condensates via a phase separation process in CRC cells. The key role of wild-type APC is to stabilize destruction complex condensates. Surprisingly, truncated APC did not affect the formation of condensates, and GSK 3ß and CK1α were unsuccessfully recruited, preventing ß-catenin phosphorylation and resulting in accumulation in the cytoplasm of CRCs. Besides, we propose that the phase separation ability of Axin participates in the nucleus translocation of ß-catenin and be incorporated and concentrated into transcriptional condensates, affecting the transcriptional activity of Wnt signaling pathway.


Subject(s)
Axin Signaling Complex , beta Catenin , Humans , Axin Signaling Complex/genetics , Axin Protein/genetics , Axin Protein/metabolism , beta Catenin/genetics , beta Catenin/metabolism , Phase Separation , Mutation/genetics , Wnt Signaling Pathway/genetics , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism
2.
Oncogene ; 40(5): 1027-1042, 2021 02.
Article in English | MEDLINE | ID: mdl-33323973

ABSTRACT

Dysregulation of the Wnt/ß-catenin signaling pathway is critically involved in gastric cancer (GC) progression. However, current Wnt pathway inhibitors being studied in preclinical or clinical settings for other cancers such as colorectal and pancreatic cancers are either too cytotoxic or insufficiently efficacious for GC. Thus, we screened new potent targets from ß-catenin destruction complex associated with GC progression from clinical samples, and found that scaffolding protein RACK1 deficiency plays a significant role in GC progression, but not APC, AXIN, and GSK3ß. Then, we identified its upstream regulator UBE2T which promotes GC progression via hyperactivating the Wnt/ß-catenin signaling pathway through the ubiquitination and degradation of RACK1 at the lysine K172, K225, and K257 residues independent of an E3 ligase. Indeed, UBE2T protein level is negatively associated with prognosis in GC patients, suggesting that UBE2T is a promising target for GC therapy. Furthermore, we identified a novel UBE2T inhibitor, M435-1279, and suggested that M435-1279 acts inhibit the Wnt/ß-catenin signaling pathway hyperactivation through blocking UBE2T-mediated degradation of RACK1, resulting in suppression of GC progression with lower cytotoxicity in the meantime. Overall, we found that increased UBE2T levels promote GC progression via the ubiquitination of RACK1 and identified a novel potent inhibitor providing a balance between growth inhibition and cytotoxicity as well, which offer a new opportunity for the specific GC patients with aberrant Wnt/ß-catenin signaling.


Subject(s)
Neoplasm Proteins/genetics , Receptors for Activated C Kinase/genetics , Stomach Neoplasms/drug therapy , Ubiquitin-Conjugating Enzymes/genetics , beta Catenin/genetics , Animals , Axin Signaling Complex/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Neoplasm Proteins/antagonists & inhibitors , Receptors for Activated C Kinase/antagonists & inhibitors , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Ubiquitination/drug effects , Wnt Signaling Pathway/genetics , Xenograft Model Antitumor Assays
3.
PLoS Biol ; 18(3): e3000647, 2020 03.
Article in English | MEDLINE | ID: mdl-32163403

ABSTRACT

Dendrite microtubules are polarized with minus-end-out orientation in Drosophila neurons. Nucleation sites concentrate at dendrite branch points, but how they localize is not known. Using Drosophila, we found that canonical Wnt signaling proteins regulate localization of the core nucleation protein γTubulin (γTub). Reduction of frizzleds (fz), arrow (low-density lipoprotein receptor-related protein [LRP] 5/6), dishevelled (dsh), casein kinase Iγ, G proteins, and Axin reduced γTub-green fluorescent protein (GFP) at branch points, and two functional readouts of dendritic nucleation confirmed a role for Wnt signaling proteins. Both dsh and Axin localized to branch points, with dsh upstream of Axin. Moreover, tethering Axin to mitochondria was sufficient to recruit ectopic γTub-GFP and increase microtubule dynamics in dendrites. At dendrite branch points, Axin and dsh colocalized with early endosomal marker Rab5, and new microtubule growth initiated at puncta marked with fz, dsh, Axin, and Rab5. We propose that in dendrites, canonical Wnt signaling proteins are housed on early endosomes and recruit nucleation sites to branch points.


Subject(s)
Dendrites/metabolism , Drosophila Proteins/metabolism , Endosomes/metabolism , Microtubules/metabolism , Wnt Proteins/metabolism , Animals , Axin Signaling Complex/genetics , Axin Signaling Complex/metabolism , Axons/metabolism , Cell Polarity , Dendrites/genetics , Drosophila , Drosophila Proteins/genetics , Endosomes/genetics , Microtubules/genetics , Mutation , Receptors, Wnt/genetics , Receptors, Wnt/metabolism , Tubulin/genetics , Tubulin/metabolism , Wnt Proteins/genetics , Wnt Signaling Pathway/genetics , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism
4.
Tohoku J Exp Med ; 249(3): 173-183, 2019 11.
Article in English | MEDLINE | ID: mdl-31723073

ABSTRACT

Colorectal cancer (CRC) is the third most common cancer and the second leading cause of death worldwide. The named "destruction complex" has a critical function in the Wnt/ß-catenin pathway regulating the level of ß-catenin in the cytoplasm and nucleus. Alterations in this complex lead to the cellular accumulation of ß-catenin, which participates in the development and progression of CRC. This study aims to determine the contribution of polymorphisms in the genes of the ß-catenin destruction complex to develop CRC, specifically adenomatous polyposis coli (APC) (rs11954856 G>T and rs459552 A>T), axis inhibition protein 1 (AXIN1) (rs9921222 C>T and rs1805105 C>T), AXIN2 (rs7224837 A>G), and dishevelled 2 (DVL2) (2074222 G>A and rs222836 C>T). Genomic DNA from 180 sporadic colorectal cancer patients and 150 healthy blood donors were analyzed. The identification of polymorphisms was made by polymerase chain reaction followed by restriction fragment length polymorphism (PCR-RFLP) methodology. Association was calculated by the odds ratio (OR) test. Increased susceptibility to CRC was associated with the polymorphic variants rs11954856 (APC), rs222836 (DVL2), and rs9921222 (AXIN1). Decreased susceptibility was associated with the polymorphisms rs459552 (APC) and 2074222 (DVL2). Association was also observed with advanced Tumor-Node-Metastasis (TNM) stages and tumor location. The haplotypes G-T in APC (rs11954856-rs459552) and A-C in DVL2 (rs2074222-rs222836) were associated with decreased risk of CRC, while the G-T haplotype in the DVL2 gene was associated with increased CRC risk. In conclusion, our results suggest that variants in the destruction complex genes may be involved in the promotion or prevention of colorectal cancer.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Axin Protein/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Dishevelled Proteins/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide/genetics , Axin Signaling Complex/genetics , Case-Control Studies , Female , Gene Frequency , Haplotypes/genetics , Humans , Male , Middle Aged , Neoplasm Staging , Wnt Signaling Pathway/genetics
5.
Dev Genes Evol ; 229(4): 89-102, 2019 07.
Article in English | MEDLINE | ID: mdl-31041506

ABSTRACT

The Wnt/beta-catenin pathway has many key roles in the development of animals, including a conserved and central role in the specification of the primary (antero-posterior) body axis. The posterior expression of Wnt ligands and the anterior expression of secreted Wnt inhibitors are known to be conserved during the larval metamorphosis of tapeworms. However, their downstream signaling components for Wnt/beta-catenin signaling have not been characterized. In this work, we have studied the core components of the beta-catenin destruction complex of the human pathogen Echinococcus multilocularis, the causative agent of alveolar echinococcosis. We focused on two Axin paralogs that are conserved in tapeworms and other flatworm parasites. Despite their divergent sequences, both Axins could robustly interact with one E. multilocularis beta-catenin paralog and limited its accumulation in a heterologous mammalian expression system. Similarly to what has been described in planarians (free-living flatworms), other beta-catenin paralogs showed limited or no interaction with either Axin and are unlikely to function as effectors in Wnt signaling. Additionally, both Axins interacted with three divergent GSK-3 paralogs that are conserved in free-living and parasitic flatworms. Axin paralogs have highly segregated expression patterns along the antero-posterior axis in the tapeworms E. multilocularis and Hymenolepis microstoma, indicating that different beta-catenin destruction complexes may operate in different regions during their larval metamorphosis.


Subject(s)
Axin Protein/genetics , Axin Signaling Complex/genetics , Echinococcus multilocularis/genetics , Glycogen Synthase Kinase 3/genetics , Helminth Proteins/genetics , Hymenolepis/genetics , beta Catenin/genetics , Amino Acid Sequence , Animals , Axin Protein/chemistry , Axin Protein/metabolism , Axin Signaling Complex/chemistry , Echinococcus multilocularis/growth & development , Echinococcus multilocularis/metabolism , Gene Expression Profiling , Glycogen Synthase Kinase 3/metabolism , Helminth Proteins/chemistry , Humans , Hymenolepis/growth & development , Hymenolepis/metabolism , Larva/metabolism , Phylogeny , Sequence Alignment , beta Catenin/metabolism
6.
Oncogene ; 38(15): 2788-2799, 2019 04.
Article in English | MEDLINE | ID: mdl-30542117

ABSTRACT

Glioblastoma (GBM) is the most aggressive brain tumor and resistant to current available therapeutics, such as radiation. To improve the clinical efficacy, it is important to understand the cellular mechanisms underlying tumor responses to radiation. Here, we investigated long-term cellular responses of human GBM cells to ionizing radiation. Comparing to the initial response within 12 hours, gene expression modulation at 7 days after radiation is markedly different. While genes related to cell cycle arrest and DNA damage responses are mostly modulated at the initial stage; immune-related genes are specifically affected as the long-term effect. This later response is associated with increased cellular senescence and inhibition of transcriptional coactivator with PDZ-binding motif (TAZ). Mechanistically, TAZ inhibition does not depend on the canonical Hippo pathway, but relies on enhanced degradation mediated by the ß-catenin destruction complex in the Wnt pathway. We further showed that depletion of TAZ by RNAi promotes radiation-induced senescence and growth arrest. Pharmacological activation of the ß-catenin destruction complex is able to promote radiation-induced TAZ inhibition and growth arrest in these tumor cells. The correlation between senescence and reduced expression of TAZ as well as ß-catenin also occurs in human gliomas treated by radiation. Collectively, these findings suggested that inhibition of TAZ is involved in radiation-induced senescence and might benefit GBM radiotherapy.


Subject(s)
Cell Cycle Checkpoints/genetics , Cell Proliferation/genetics , Cellular Senescence/genetics , Glioma/genetics , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Axin Signaling Complex/genetics , Brain Neoplasms/genetics , Brain Neoplasms/radiotherapy , Cell Cycle Checkpoints/radiation effects , Cell Line, Tumor , Gene Expression/genetics , Gene Expression/radiation effects , Glioblastoma/genetics , Glioblastoma/radiotherapy , Glioma/radiotherapy , Humans , Radiation, Ionizing , Radiotherapy/methods , Trans-Activators , Transcriptional Coactivator with PDZ-Binding Motif Proteins , beta Catenin/genetics
7.
Mol Cell Biol ; 38(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30181396

ABSTRACT

The ubiquitous transcription factor specificity protein 1 (SP1) is heavily modified posttranslationally. These modifications are critical for switching its functions and modulation of its transcriptional activity and DNA binding and stability. However, the mechanism governing the stability of SP1 by cellular signaling pathways is not well understood. Here, we provide biochemical and functional evidence that SP1 is an integral part of the Wnt signaling pathway. We identified a phosphodegron motif in SP1 that is specific to mammals. In the absence of Wnt signaling, glycogen synthase kinase 3ß (GSK3ß)-mediated phosphorylation and ß-TrCP E3 ubiquitin ligase-mediated ubiquitination are required to induce SP1 degradation. When Wnt signaling is on, SP1 is stabilized in a ß-catenin-dependent manner. SP1 directly interacts with ß-catenin, and Wnt signaling induces the stabilization of SP1 by impeding its interaction with ß-TrCP and axin1, components of the destruction complex. Wnt signaling suppresses ubiquitination and subsequent proteosomal degradation of SP1. Furthermore, SP1 regulates Wnt-dependent stability of ß-catenin and their mutual stabilization is critical for target gene expression, suggesting a feedback mechanism. Upon stabilization, SP1 and ß-catenin cooccupy the promoters of TCFL2/ß-catenin target genes. Collectively, this study uncovers a direct link between SP1 and ß-catenin in the Wnt signaling pathway.


Subject(s)
Axin Signaling Complex/genetics , Sp1 Transcription Factor/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Amino Acid Sequence , Animals , Cell Line , Cell Line, Tumor , Glycogen Synthase Kinase 3 beta/genetics , HCT116 Cells , HEK293 Cells , Humans , Phosphorylation/genetics , Sequence Alignment , Transcription, Genetic/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , beta-Transducin Repeat-Containing Proteins/genetics
8.
PLoS Genet ; 14(4): e1007339, 2018 04.
Article in English | MEDLINE | ID: mdl-29641560

ABSTRACT

Wnt signaling provides a paradigm for cell-cell signals that regulate embryonic development and stem cell homeostasis and are inappropriately activated in cancers. The tumor suppressors APC and Axin form the core of the multiprotein destruction complex, which targets the Wnt-effector beta-catenin for phosphorylation, ubiquitination and destruction. Based on earlier work, we hypothesize that the destruction complex is a supramolecular entity that self-assembles by Axin and APC polymerization, and that regulating assembly and stability of the destruction complex underlie its function. We tested this hypothesis in Drosophila embryos, a premier model of Wnt signaling. Combining biochemistry, genetic tools to manipulate Axin and APC2 levels, advanced imaging and molecule counting, we defined destruction complex assembly, stoichiometry, and localization in vivo, and its downregulation in response to Wnt signaling. Our findings challenge and revise current models of destruction complex function. Endogenous Axin and APC2 proteins and their antagonist Dishevelled accumulate at roughly similar levels, suggesting competition for binding may be critical. By expressing Axin:GFP at near endogenous levels we found that in the absence of Wnt signals, Axin and APC2 co-assemble into large cytoplasmic complexes containing tens to hundreds of Axin proteins. Wnt signals trigger recruitment of these to the membrane, while cytoplasmic Axin levels increase, suggesting altered assembly/disassembly. Glycogen synthase kinase3 regulates destruction complex recruitment to the membrane and release of Armadillo/beta-catenin from the destruction complex. Manipulating Axin or APC2 levels had no effect on destruction complex activity when Wnt signals were absent, but, surprisingly, had opposite effects on the destruction complex when Wnt signals were present. Elevating Axin made the complex more resistant to inactivation, while elevating APC2 levels enhanced inactivation. Our data suggest both absolute levels and the ratio of these two core components affect destruction complex function, supporting models in which competition among Axin partners determines destruction complex activity.


Subject(s)
Armadillo Domain Proteins/metabolism , Axin Signaling Complex/metabolism , Drosophila Proteins/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway , Animals , Animals, Genetically Modified , Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome/chemistry , Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Armadillo Domain Proteins/chemistry , Armadillo Domain Proteins/genetics , Axin Protein/chemistry , Axin Protein/genetics , Axin Protein/metabolism , Axin Signaling Complex/chemistry , Axin Signaling Complex/genetics , Cell Line , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Proteolysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription, Genetic , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
9.
J Proteome Res ; 16(10): 3576-3584, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28810742

ABSTRACT

Wnt/ß-catenin signaling plays important roles in both ontogenesis and development. In the absence of a Wnt stimulus, ß-catenin is degraded by a multiprotein "destruction complex" that includes Axin, APC, GSK3B, and FBXW11. Although the key molecules required for transducing Wnt signals have been identified, a quantitative understanding of this pathway has been lacking. Here, we calculated the absolute number of ß-catenin destruction complexes by absolute protein quantification using LC-MS/MS. Similar amounts of destruction complex-constituting proteins and ß-catenin interacted, and the number of destruction complexes was calculated to be about 1468 molecules/cell. We demonstrated that the calculated number of destruction complexes was valid for control of the ß-catenin destruction rate under steady-state conditions. Interestingly, APC had the minimum expression level among the destruction complex components at about 2233 molecules/cell, and this number approximately corresponded to the calculated number of destruction complexes. Decreased APC expression by siRNA transfection decreased the number of destruction complexes, resulting in ß-catenin accumulation and stimulation of the transcriptional activity of T-cell factor. Taken together, our results suggest that the amount of APC expression is the rate-limiting factor for the constitution of ß-catenin destruction complexes.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Axin Signaling Complex/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Axin Protein/genetics , Axin Signaling Complex/chemistry , Axin Signaling Complex/metabolism , Gene Expression Regulation/genetics , Glycogen Synthase Kinase 3 beta/genetics , HCT116 Cells , Humans , Phosphorylation , RNA, Small Interfering/genetics , Ubiquitin-Protein Ligases/genetics , beta Catenin/isolation & purification , beta-Transducin Repeat-Containing Proteins/genetics
10.
Cancer Med ; 3(1): 36-46, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24403055

ABSTRACT

Wnt/ß-catenin is a major regulator of stem cell self-renewal and differentiation and this signaling pathway is aberrantly activated in a several cancers, including osteosarcoma (OS). Attenuation of Wnt/ß-catenin activity by tankyrase inhibitors is an appealing strategy in treatment of OS. The efficacy of the tankyrase inhibitor JW74 was evaluated in three OS cell lines (KPD, U2OS, and SaOS-2) both at the molecular and functional level. At the molecular level, JW74 induces stabilization of AXIN2, a key component of the ß-catenin destruction complex, resulting in reduced levels of nuclear ß-catenin. At the functional level, JW74 induces reduced cell growth in all three tested cell lines, in part due to a delay in cell cycle progression and in part due to an induction of caspase-3-mediated apoptosis. Furthermore, JW74 induces differentiation in U2OS cells, which under standard conditions are resistant to osteogenic differentiation. JW74 also enhances differentiation of OS cell lines, which do not harbor a differentiation block. Interestingly, microRNAs (miRNAs) of the let-7 family, which are known tumor suppressors and inducers of differentiation, are significantly upregulated following treatment with JW74. We demonstrate for the first time that tankyrase inhibition triggers reduced cell growth and differentiation of OS cells. This may in part be due to an induction of let-7 miRNA. The presented data open for novel therapeutic strategies in the treatment of malignant OS.


Subject(s)
Bone Neoplasms/drug therapy , Cell Differentiation/drug effects , Osteosarcoma/drug therapy , Oxadiazoles/administration & dosage , Tankyrases/genetics , Triazoles/administration & dosage , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Axin Signaling Complex/genetics , Bone Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Osteogenesis/genetics , Osteosarcoma/genetics , Osteosarcoma/pathology , Tankyrases/antagonists & inhibitors , Wnt Signaling Pathway/genetics
11.
Gastroenterology ; 142(4): 812-823.e15, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22240482

ABSTRACT

BACKGROUND & AIMS: Dysregulation of Wnt signaling has been involved in gastric tumorigenesis by mechanisms that are not fully understood. The receptor for activated protein kinase C (RACK1, GNB2L1) is involved in development of different tumor types, but its expression and function have not been investigated in gastric tumors. METHODS: We analyzed expression of RACK1 in gastric tumor samples and their matched normal tissues from 116 patients using immunohistochemistry. Effects of knockdown with small interfering RNAs or overexpression of RACK1 in gastric cancer cell lines were evaluated in cell growth and tumor xenograft. RACK1 signaling pathways were investigated in cells and zebrafish embryos using immunoblot, immunoprecipitation, microinjection, and in situ hybridization assays. RESULTS: Expression of RACK1 was reduced in gastric tumor samples and correlated with depth of tumor infiltration and poor differentiation. Knockdown of RACK1 in gastric cancer cells accelerated their anchorage-independent proliferation in soft agar, whereas overexpression of RACK1 reduced their tumorigenicity in nude mice. RACK1 formed a complex with glycogen synthase kinase Gsk3ß and Axin to promote the interaction between Gsk3ß and ß-catenin and thereby stabilized the ß-catenin destruction complex. On stimulation of Wnt3a, RACK1 repressed Wnt signaling by inhibiting recruitment of Axin by Dishevelled 2 (Dvl2). Moreover, there was an inverse correlation between expression of RACK1 and localization of ß-catenin to the cytoplasm/nucleus in human gastric tumor samples. CONCLUSIONS: RACK1 negatively regulates Wnt signaling pathway by stabilizing the ß-catenin destruction complex and act as a tumor suppressor in gastric cancer cells.


Subject(s)
Axin Signaling Complex/metabolism , GTP-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Receptors, Cell Surface/metabolism , Stomach Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Wnt Signaling Pathway , Adaptor Proteins, Signal Transducing/metabolism , Animals , Axin Signaling Complex/genetics , Case-Control Studies , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Dishevelled Proteins , Female , GTP-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasm Transplantation , Phosphoproteins/metabolism , RNA Interference , Receptors for Activated C Kinase , Receptors, Cell Surface/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/prevention & control , Time Factors , Transfection , Tumor Suppressor Proteins/genetics , Wnt Signaling Pathway/genetics , Wnt3A Protein/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , beta Catenin/metabolism
13.
Open Biol ; 1(3): 110013, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22645652

ABSTRACT

Most cases of colorectal cancer are linked to mutational inactivation of the Adenomatous polyposis coli (APC) tumour suppressor. APC downregulates Wnt signalling by enabling Axin to promote the degradation of the Wnt signalling effector ß-catenin (Armadillo in flies). This depends on Axin's DIX domain whose polymerization allows it to form dynamic protein assemblies ('degradasomes'). Axin is inactivated upon Wnt signalling, by heteropolymerization with the DIX domain of Dishevelled, which recruits it into membrane-associated 'signalosomes'. How APC promotes Axin's function is unclear, especially as it has been reported that APC's function can be bypassed by overexpression of Axin. Examining apc null mutant Drosophila tissues, we discovered that APC is required for Axin degradasome assembly, itself essential for Armadillo downregulation. Degradasome assembly is also attenuated in APC mutant cancer cells. Notably, Axin becomes prone to Dishevelled-dependent plasma membrane recruitment in the absence of APC, indicating a crucial role of APC in opposing the interaction of Axin with Dishevelled. Indeed, co-expression experiments reveal that APC displaces Dishevelled from Axin assemblies, promoting degradasome over signalosome formation in the absence of Wnts. APC thus empowers Axin to function in two ways-by enabling its DIX-dependent self-assembly, and by opposing its DIX-dependent copolymerization with Dishevelled and consequent inactivation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Axin Protein/metabolism , Axin Signaling Complex/metabolism , Cytoskeletal Proteins/metabolism , Drosophila Proteins/metabolism , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/metabolism , Animals , Animals, Genetically Modified , Axin Protein/chemistry , Axin Protein/genetics , Axin Signaling Complex/chemistry , Axin Signaling Complex/genetics , Cell Line, Tumor , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Dishevelled Proteins , Drosophila/embryology , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Endoribonucleases/metabolism , Genes, APC , Genes, Insect , Humans , Multienzyme Complexes/metabolism , Mutation , Phosphoproteins/chemistry , Phosphoproteins/genetics , Polyribonucleotide Nucleotidyltransferase/metabolism , Protein Interaction Domains and Motifs , RNA Helicases/metabolism , Wnt Signaling Pathway
SELECTION OF CITATIONS
SEARCH DETAIL
...