Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
J Cell Sci ; 136(20)2023 10 15.
Article En | MEDLINE | ID: mdl-37721093

Axin (also known as AXIN1) is a central negative regulator of the proto-oncogenic Wnt/ß-catenin signaling pathway, as axin condensates provide a scaffold for the assembly of a multiprotein complex degrading ß-catenin. Axin, in turn, is degraded through tankyrase. Consequently, tankyrase small-molecule inhibitors block Wnt signaling by stabilizing axin, revealing potential for cancer therapy. Here, we discovered that axin is phosphorylated by casein kinase 1 alpha 1 (CSNK1A1, also known as CK1α) at an N-terminal casein kinase 1 consensus motif, and that this phosphorylation is antagonized by the catalytic subunit alpha of protein phosphatase 1 (PPP1CA, hereafter referred to as PP1). Axin condensates promoted phosphorylation by enriching CK1α over PP1. Importantly, the phosphorylation took place within the tankyrase-binding site, electrostatically and/or sterically hindering axin-tankyrase interaction, and counteracting tankyrase-mediated degradation of axin. Thus, the presented data propose a novel mechanism regulating axin stability, with implications for Wnt signaling, cancer therapy and self-organization of biomolecular condensates.


Neoplasms , Tankyrases , Humans , Axin Protein/metabolism , Phosphorylation , Tankyrases/metabolism , Biomolecular Condensates , beta Catenin/metabolism , Wnt Signaling Pathway
2.
Nat Commun ; 13(1): 674, 2022 02 03.
Article En | MEDLINE | ID: mdl-35115535

Conductin/axin2 is a scaffold protein negatively regulating the pro-proliferative Wnt/ß-catenin signaling pathway. Accumulation of scaffold proteins in condensates frequently increases their activity, but whether condensation contributes to Wnt pathway inhibition by conductin remains unclear. Here, we show that the Gαi2 subunit of trimeric G-proteins induces conductin condensation by targeting a polymerization-inhibiting aggregon in its RGS domain, thereby promoting conductin-mediated ß-catenin degradation. Consistently, transient Gαi2 expression inhibited, whereas knockdown activated Wnt signaling via conductin. Colorectal cancers appear to evade Gαi2-induced Wnt pathway suppression by decreased Gαi2 expression and inactivating mutations, associated with shorter patient survival. Notably, the Gαi2-activating drug guanabenz inhibited Wnt signaling via conductin, consequently reducing colorectal cancer growth in vitro and in mouse models. In summary, we demonstrate Wnt pathway inhibition via Gαi2-triggered conductin condensation, suggesting a tumor suppressor function for Gαi2 in colorectal cancer, and pointing to the FDA-approved drug guanabenz for targeted cancer therapy.


Axin Protein/genetics , Colorectal Neoplasms/genetics , GTP-Binding Protein alpha Subunit, Gi2/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Adrenergic alpha-2 Receptor Agonists/pharmacology , Animals , Axin Protein/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Gene Expression Regulation, Neoplastic , Guanabenz/pharmacology , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mutation , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays/methods , beta Catenin/metabolism
3.
Sci Rep ; 10(1): 8323, 2020 05 20.
Article En | MEDLINE | ID: mdl-32433485

Viral infections trigger host innate immune responses, characterized by the production of type-I interferons (IFN) including IFNß. IFNß induces cellular antiviral defense mechanisms and thereby contributes to pathogen clearance. Accumulating evidence suggests that mitochondria constitute a crucial platform for the induction of antiviral immunity. Here we demonstrate that the mitochondrial protein phosphoglycerate mutase family member 5 (PGAM5) is important for the antiviral cellular response. Following challenge of HeLa cells with the dsRNA-analog poly(I:C), PGAM5 oligomers and high levels of PGAM5 were found in mitochondrial aggregates. Using immunoprecipitation, a direct interaction of PGAM5 with the mitochondrial antiviral-signaling protein (MAVS) was demonstrated. In addition, PGAM5 deficient cells showed diminished expression of IFNß and IFNß target genes as compared to WT cells. Moreover, PGAM5 deficient mouse embryonic fibroblasts (MEFs) exhibited decreased phosphorylation levels of IRF3 and TBK1 when challenged with poly(I:C) intracellularly. Finally, PGAM5 deficient MEFs, upon infection with vesicular stomatitis virus (VSV), revealed diminished IFNß expression and increased VSV replication. Collectively, our study highlights PGAM5 as an important regulator for IFNß production mediated via the TBK1/IRF3 signaling pathway in response to viral infection.


Adaptor Proteins, Signal Transducing/metabolism , Mitochondrial Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Vesicular stomatitis Indiana virus/immunology , Animals , Cells, Cultured , Fibroblasts/virology , HeLa Cells , Humans , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Mice , Mitochondrial Proteins/immunology , Phosphoprotein Phosphatases/immunology , Poly I-C/immunology , Protein Serine-Threonine Kinases/metabolism , Rhabdoviridae Infections/immunology , Signal Transduction , Virus Replication/immunology
4.
Nat Commun ; 10(1): 4251, 2019 09 18.
Article En | MEDLINE | ID: mdl-31534175

The paralogous scaffold proteins axin and conductin/axin2 are key factors in the negative regulation of the Wnt pathway transcription factor ß-catenin, thereby representing interesting targets for signaling regulation. Polymerization of axin proteins is essential for their activity in suppressing Wnt/ß-catenin signaling. Notably, conductin shows less polymerization and lower activity than axin. By domain swapping between axin and conductin we here identify an aggregation site in the conductin RGS domain which prevents conductin polymerization. Induction of conductin polymerization by point mutations of this aggregon results in enhanced inhibition of Wnt/ß-catenin signaling. Importantly, we identify a short peptide which induces conductin polymerization via masking the aggregon, thereby enhancing ß-catenin degradation, inhibiting ß-catenin-dependent transcription and repressing growth of colorectal cancer cells. Our study reveals a mechanism for regulating signaling pathways via the polymerization status of scaffold proteins and suggests a strategy for targeted colorectal cancer therapy.


Axin Protein/metabolism , Colorectal Neoplasms/pathology , Wnt Signaling Pathway/genetics , beta Catenin/metabolism , Axin Protein/genetics , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/genetics , Gene Knockout Techniques , HEK293 Cells , Humans , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology
5.
Oncotarget ; 9(74): 33982-33994, 2018 Sep 21.
Article En | MEDLINE | ID: mdl-30338040

The naturally occurring isothiocyanate sulforaphane (SFN) from cruciferous vegetables is associated with growth inhibition of various cancer types, including colorectal cancer. Colorectal cancer is most frequently driven by hyperactive Wnt/ß-catenin signaling. Here, we show that SFN treatment reduced growth of three unrelated colorectal cancer cell lines (SW480, DLD1 and HCT116) via induction of cell death and inhibition of proliferation. Importantly, SFN inhibits Wnt/ß-catenin signaling in colorectal cancer cells as shown by inhibition of ß-catenin-dependent luciferase reporters and repression of ß-catenin target genes (AXIN2, LGR5). SFN inhibits Wnt signaling downstream of ß-catenin degradation and induces the formation of nuclear ß-catenin structures associated with closed chromatin. Co-expression of the transcription factors LEF1 or TCF4 prevented formation of these structures and rescued inhibition of Wnt/ß-catenin signaling by SFN. Our findings provide a molecular basis explaining SFN effects in colorectal cancer cells and underline its potential for prevention and therapy of colorectal cancer.

6.
Mol Cell Oncol ; 5(3): e1458015, 2018.
Article En | MEDLINE | ID: mdl-30250905

Cellular abundance of mitochondria is dynamically regulated. We could recently show that dysfunctional mitochondria release the phosphatase PGAM family member 5 (PGAM5) into the cytosol, where it interacts with the Wnt signaling-component AXIN1 and dephosphorylates AXIN1-bound ß-catenin (CTNNB1) thereby activating Wnt/ß-catenin signaling. Because Wnt/ß-catenin signaling induces mitochondrial biogenesis dysfunctional mitochondria trigger their own replacement by releasing PGAM5.

8.
J Cell Biol ; 217(4): 1383-1394, 2018 04 02.
Article En | MEDLINE | ID: mdl-29438981

Mitochondrial abundance is dynamically regulated and was previously shown to be increased by Wnt/ß-catenin signaling. Pgam5 is a mitochondrial phosphatase which is cleaved by the rhomboid protease presenilin-associated rhomboid-like protein (PARL) and released from membranes after mitochondrial stress. In this study, we show that Pgam5 interacts with the Wnt pathway component axin in the cytosol, blocks axin-mediated ß-catenin degradation, and increases ß-catenin levels and ß-catenin-dependent transcription. Pgam5 stabilized ß-catenin by inducing its dephosphorylation in an axin-dependent manner. Mitochondrial stress triggered by carbonyl cyanide m-chlorophenyl hydrazone (CCCP) treatment led to cytosolic release of endogenous Pgam5 and subsequent dephosphorylation of ß-catenin, which was strongly diminished in Pgam5 and PARL knockout cells. Similarly, hypoxic stress generated cytosolic Pgam5 and led to stabilization of ß-catenin, which was abolished by Pgam5 knockout. Cells stably expressing cytosolic Pgam5 exhibit elevated ß-catenin levels and increased mitochondrial numbers. Our study reveals a novel mechanism by which damaged mitochondria might induce replenishment of the mitochondrial pool by cell-intrinsic activation of Wnt signaling via the Pgam5-ß-catenin axis.


Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Organelle Biogenesis , Phosphoprotein Phosphatases/metabolism , Wnt Signaling Pathway , Animals , Axin Protein/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Phosphoprotein Phosphatases/genetics , Phosphorylation , Protein Binding , Protein Stability , Proteolysis , Proton Ionophores/toxicity , Transcription, Genetic , Transcriptional Activation , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
9.
Development ; 144(12): 2234-2247, 2017 06 15.
Article En | MEDLINE | ID: mdl-28506997

The scaffold protein Dishevelled is a central intracellular component of Wnt signaling pathways. Various kinases have been described that regulate and modulate Wnt signaling through phosphorylation of Dishevelled. However, besides general protein phosphatases 1 and 2 (PP1 and PP2), no specific protein phosphatases have been identified. Here, we report on the identification and functional characterization of the protein phosphatase Pgam5 in vitro and in vivo in Xenopus Pgam5 is a novel antagonist of Wnt/ß-Catenin signaling in human cells and Xenopus embryogenesis. In early development, Pgam5 is essential for head formation, and for establishing and maintaining the Wnt/ß-Catenin signaling gradient that patterns the anterior-posterior body axis. Inhibition of Wnt/ß-Catenin signaling and developmental function depend on Pgam5 phosphatase activity. We show that Pgam5 interacts with Dishevelled2 and that Dishevelled2 is a substrate of Pgam5. Pgam5 mediates a marked decrease in Dishevelled2 phosphorylation in the cytoplasm and in the nucleus, as well as decreased interaction between Dishevelled2, Tcf1 and ß-Catenin, indicating that Pgam5 regulates Dishevelled function upstream and downstream of ß-Catenin stabilization.


Body Patterning/physiology , Phosphoprotein Phosphatases/metabolism , Wnt Signaling Pathway , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Xenopus laevis/metabolism , beta Catenin/metabolism , Amino Acid Sequence , Animals , Body Patterning/genetics , Dishevelled Proteins/genetics , Dishevelled Proteins/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/genetics , Phosphoproteins , Sequence Homology, Amino Acid , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/genetics , Xenopus laevis/genetics , beta Catenin/genetics , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism
10.
J Cell Sci ; 128(1): 33-9, 2015 Jan 01.
Article En | MEDLINE | ID: mdl-25380820

Axin and conductin (also known as axin2) are structurally related inhibitors of Wnt/ß-catenin signalling that promote degradation of ß-catenin. Whereas axin is constitutively expressed, conductin is a Wnt target gene implicated in Wnt negative-feedback regulation. Here, we show that axin and conductin differ in their functional interaction with the upstream Wnt pathway component Dvl. Conductin shows reduced binding to Dvl2 compared to axin, and degradation of ß-catenin by conductin is only poorly blocked by Dvl2. We propose that insensitivity to Dvl is an important feature of the role of conductin as a negative-feedback regulator of Wnt signalling.


Adaptor Proteins, Signal Transducing/metabolism , Axin Protein/metabolism , Phosphoproteins/metabolism , Wnt Signaling Pathway/physiology , Adaptor Proteins, Signal Transducing/genetics , Axin Protein/genetics , Dishevelled Proteins , HEK293 Cells , Humans , Phosphoproteins/genetics
11.
FEBS J ; 281(3): 787-801, 2014 Feb.
Article En | MEDLINE | ID: mdl-24251807

The adenomatous polyposis coli (APC) membrane recruitment (Amer) family proteins Amer1/Wilms tumour gene on the X chromosome and Amer2 are binding partners of the APC tumour suppressor protein, and act as negative regulators in the Wnt signalling cascade. So far, nothing has been known about the third member of the family, Amer3. Here we show that Amer3 binds to the armadillo repeat domain of APC, similarly to Amer1 and Amer2. Amer3 also binds to the Wnt pathway regulator conductin/axin2. Furthermore, we identified Amer1 as binding partner of Amer3. Whereas Amer1 and Amer2 are linked to the plasma membrane by an N-terminal membrane localization domain, Amer3 lacks this domain. Amer3 localizes to the cytoplasm and nucleus of epithelial cells, and this is dependent on specific nuclear import and export sequences. Functionally, exogenous Amer3 enhances the expression of a ß-catenin/T-cell factor-dependent reporter gene, and knockdown of endogenous Amer3 reduces Wnt target gene expression in colorectal cancer cells. Thus, Amer3 acts as an activator of Wnt signalling, in contrast to Amer1 and Amer2, which are inhibitors, suggesting a nonredundant role of Amer proteins in the regulation of this pathway. Our data, together with those of previous studies, provide a comprehensive picture of similarities and differences within the Amer protein family.


Adaptor Proteins, Signal Transducing/metabolism , Adenomatous Polyposis Coli Protein/metabolism , Adenomatous Polyposis Coli/metabolism , Cell Nucleus/metabolism , Colorectal Neoplasms/metabolism , Cytoplasm/metabolism , Tumor Suppressor Proteins/metabolism , Up-Regulation , Wnt Signaling Pathway , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Adenomatous Polyposis Coli Protein/chemistry , Adenomatous Polyposis Coli Protein/genetics , Armadillo Domain Proteins/chemistry , Armadillo Domain Proteins/metabolism , Axin Protein/metabolism , Cell Line, Tumor , Colorectal Neoplasms/pathology , HEK293 Cells , Humans , Mutant Proteins , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Organ Specificity , Protein Interaction Domains and Motifs , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Sorting Signals , Protein Transport , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
12.
EMBO Rep ; 13(4): 347-54, 2012 Apr.
Article En | MEDLINE | ID: mdl-22322943

Wnt/ß-catenin signalling regulates cell proliferation by modulating the cell cycle and is negatively regulated by conductin/axin2/axil. We show that conductin levels peak at G2/M followed by a rapid decline during return to G1. In line with this, Wnt/ß-catenin target genes are low at G2/M and high at G1/S, and ß-catenin phosphorylation oscillates during the cell cycle in a conductin-dependent manner. Conductin is degraded by the anaphase-promoting complex/cyclosome cofactor CDC20. Knockdown of CDC20 blocks Wnt signalling through conductin. CDC20-resistant conductin inhibits Wnt signalling and attenuates colony formation of colorectal cancer cells. We propose that CDC20-mediated degradation of conductin regulates Wnt/ß-catenin signalling for maximal activity during G1/S.


Axin Protein/metabolism , Cell Cycle Checkpoints , Cell Cycle Proteins/metabolism , Wnt Signaling Pathway , Amino Acid Sequence , Animals , Axin Protein/chemistry , Cdc20 Proteins , Cell Line, Tumor , Conserved Sequence , Humans , Mice , Mitosis , Molecular Sequence Data , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Stability , Protein Structure, Tertiary , Proteolysis , Rats , beta Catenin/metabolism
13.
Expert Opin Ther Targets ; 12(7): 845-53, 2008 Jul.
Article En | MEDLINE | ID: mdl-18554153

BACKGROUND: Loss of erythrocyte membrane protein band 4.1-like 3 (EPB41L3; aliases: protein 4.1B, differentially expressed in adenocarcinoma of the lung-1 (Dal-1)) expression has been implicated in tumor progression. OBJECTIVE: To evaluate literature describing the role of EPB41L3 in tumorigenesis and metastasis, and to consider whether targeting this gene would be useful in the treatment of prostate cancer. METHODS: A literature review of studies describing EPB41L3 and its aliases was conducted. Online databases (NCBI, SwissProt) were also interrogated to collect further data. RESULTS/CONCLUSION: A growing body of evidence supports a role for loss of EPB41L3 in tumor progression, including in prostate cancer. Therapeutic strategies that could be harnessed to upregulate EPB41L3 gene expression in prostate cancer cells are currently being developed.


Antineoplastic Agents/pharmacology , Membrane Proteins/drug effects , Prostatic Neoplasms/physiopathology , Tumor Suppressor Proteins/drug effects , Animals , Disease Progression , Drug Delivery Systems , Gene Expression Regulation/drug effects , Humans , Male , Membrane Proteins/metabolism , Microfilament Proteins , Neoplasm Metastasis/physiopathology , Prostatic Neoplasms/drug therapy , Tumor Suppressor Proteins/metabolism
...