Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Nat Rev Mol Cell Biol ; 17(1): 55-64, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26580716

ABSTRACT

ß-catenin is widely regarded as the primary transducer of canonical WNT signals to the nucleus. In most vertebrates, there are eight additional catenins that are structurally related to ß-catenin, and three α-catenin genes encoding actin-binding proteins that are structurally related to vinculin. Although these catenins were initially identified in association with cadherins at cell-cell junctions, more recent evidence suggests that the majority of catenins also localize to the nucleus and regulate gene expression. Moreover, the number of catenins reported to be responsive to canonical WNT signals is increasing. Here, we posit that multiple catenins form a functional network in the nucleus, possibly engaging in conserved protein-protein interactions that are currently better characterized in the context of actin-based cell junctions.


Subject(s)
Cell Nucleus/metabolism , beta Catenin/metabolism , Animals , Armadillo Domain Proteins/chemistry , Armadillo Domain Proteins/metabolism , Humans , Models, Biological , Signal Transduction , Wnt Proteins/metabolism , beta Catenin/chemistry
2.
Biochem Biophys Res Commun ; 563: 31-39, 2021 07 23.
Article in English | MEDLINE | ID: mdl-34058472

ABSTRACT

Vertebrate beta-catenin plays a key role as a transducer of canonical-Wnt signals. We earlier reported that, similar to beta-catenin, the cytoplasmic signaling pool of p120-catenin-isoform1 is stabilized in response to canonical-Wnt signals. To obtain a yet broader view of the Wnt-pathway's impact upon catenin proteins, we focused upon plakophilin3 (plakophilin-3; Pkp3) as a representative of the plakophilin-catenin subfamily. Promoting tissue integrity, the plakophilins assist in linking desmosomal cadherins to intermediate filaments at desmosome junctions, and in common with other catenins they perform additional functions including in the nucleus. In this report, we test whether canonical-Wnt pathway components modulate Pkp3 protein levels. We find that in common with beta-catenin and p120-catenin-isoform1, Pkp3 is stabilized in the presence of a Wnt-ligand or a dominant-active form of the LRP6 receptor. Pkp3's levels are conversely lowered upon expressing destruction-complex components such as GSK3ß and Axin, and in further likeness to beta-catenin and p120-isoform1, Pkp3 associates with GSK3beta and Axin. Finally, we note that Pkp3-catenin trans-localizes into the nucleus in response to Wnt-ligand and its exogenous expression stimulates an accepted Wnt reporter. These findings fit an expanded model where context-dependent Wnt-signals or pathway components modulate Pkp3-catenin levels. Future studies will be needed to assess potential gene regulatory, cell adhesive, or cytoskeletal effects.


Subject(s)
Plakophilins/metabolism , Animals , Cells, Cultured , Humans , Wnt Signaling Pathway , Xenopus laevis
3.
Mol Cell ; 52(2): 193-205, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24055345

ABSTRACT

Fine control of Wnt signaling is essential for various cellular and developmental decision-making processes. However, deregulation of Wnt signaling leads to pathological consequences, one of which is cancer. Here, we identify a function of PAF, a component of translesion DNA synthesis, in modulating Wnt signaling. PAF is specifically overexpressed in colon cancer cells and intestinal stem cells and is required for colon cancer cell proliferation. In Xenopus laevis, ventrovegetal expression of PAF hyperactivates Wnt signaling, developing a secondary axis with ß-catenin target gene upregulation. Upon Wnt signaling activation, PAF dissociates from PCNA and binds directly to ß-catenin. Then, PAF recruits EZH2 to the ß-catenin transcriptional complex and specifically enhances Wnt target gene transactivation, independently of EZH2's methyltransferase activity. In mice, conditional expression of PAF induces intestinal neoplasia via Wnt signaling hyperactivation. Our studies reveal an unexpected role of PAF in regulating Wnt signaling and propose a regulatory mechanism of Wnt signaling during tumorigenesis.


Subject(s)
Carrier Proteins/metabolism , Polycomb Repressive Complex 2/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA-Binding Proteins , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation, Developmental , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polycomb Repressive Complex 2/genetics , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Xenopus laevis , beta Catenin/genetics
4.
Biochim Biophys Acta ; 1863(1): 102-14, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26477567

ABSTRACT

P120-catenin is essential to vertebrate development, modulating cadherin and small-GTPase functions, and growing evidence points also to roles in the nucleus. A complexity in addressing p120-catenin's functions is its many isoforms, including optional splicing events, alternative points of translational initiation, and secondary modifications. In this review, we focus upon how choices in the initiation of protein translation, or the earlier splicing of the RNA transcript, relates to primary sequences that harbor established or putative regulatory phosphorylation sites. While certain p120 phosphorylation events arise via known kinases/phosphatases and have defined outcomes, in most cases the functional consequences are still to be established. In this review, we provide examples of p120-isoforms as they relate to phosphorylation events, and thereby to isoform dependent protein-protein associations and downstream functions. We also provide a view of upstream pathways that determine p120's phosphorylation state, and that have an impact upon development and disease. Because other members of the p120 subfamily undergo similar processing and phosphorylation, as well as related catenins of the plakophilin subfamily, what is learned regarding p120 will by extension have wide relevance in vertebrates.


Subject(s)
Catenins/metabolism , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Animals , Catenins/genetics , Cell Nucleus/genetics , Cell Transformation, Neoplastic/genetics , Humans , Neoplasm Proteins/genetics , Neoplasms/genetics , Phosphorylation/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Delta Catenin
5.
J Cell Sci ; 127(Pt 18): 4037-51, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25074806

ABSTRACT

Although the canonical Wnt pathway and ß-catenin have been extensively studied, less is known about the role of p120-catenin (also known as δ1-catenin) in the nuclear compartment. Here, we report that p120-catenin binds and negatively regulates REST and CoREST (also known as Rcor1), a repressive transcriptional complex that has diverse developmental and pathological roles. Using mouse embryonic stem cells (mESCs), mammalian cell lines, Xenopus embryos and in vitro systems, we find that p120-catenin directly binds the REST-CoREST complex, displacing it from established gene targets to permit their transcriptional activation. Importantly, p120-catenin levels further modulate the mRNA and protein levels of Oct4 (also known as POU5F1), Nanog and Sox2, and have an impact upon the differentiation of mESCs towards neural fates. In assessing potential upstream inputs to this new p120-catenin-REST-CoREST pathway, REST gene targets were found to respond to the level of E-cadherin, with evidence suggesting that p120-catenin transduces signals between E-cadherin and the nucleus. In summary, we provide the first evidence for a direct upstream modulator and/or pathway regulating REST-CoREST, and reveal a substantial role for p120-catenin in the modulation of stem cell differentiation.


Subject(s)
Catenins/metabolism , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Repressor Proteins/metabolism , Animals , Catenins/genetics , Co-Repressor Proteins , Humans , Mice , Nerve Tissue Proteins/genetics , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Repressor Proteins/genetics , Xenopus laevis , Delta Catenin
6.
Proc Natl Acad Sci U S A ; 110(45): 18042-51, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24058167

ABSTRACT

Mammalian organs, including the lung and kidney, often adopt a branched structure to achieve high efficiency and capacity of their physiological functions. Formation of a functional lung requires two developmental processes: branching morphogenesis, which builds a tree-like tubular network, and alveolar differentiation, which generates specialized epithelial cells for gas exchange. Much progress has been made to understand each of the two processes individually; however, it is not clear whether the two processes are coordinated and how they are deployed at the correct time and location. Here we show that an epithelial branching morphogenesis program antagonizes alveolar differentiation in the mouse lung. We find a negative correlation between branching morphogenesis and alveolar differentiation temporally, spatially, and evolutionarily. Gain-of-function experiments show that hyperactive small GTPase Kras expands the branching program and also suppresses molecular and cellular differentiation of alveolar cells. Loss-of-function experiments show that SRY-box containing gene 9 (Sox9) functions downstream of Fibroblast growth factor (Fgf)/Kras to promote branching and also suppresses premature initiation of alveolar differentiation. We thus propose that lung epithelial progenitors continuously balance between branching morphogenesis and alveolar differentiation, and such a balance is mediated by dual-function regulators, including Kras and Sox9. The resulting temporal delay of differentiation by the branching program may provide new insights to lung immaturity in preterm neonates and the increase in organ complexity during evolution.


Subject(s)
Cell Differentiation/physiology , Lung/embryology , Morphogenesis/physiology , Proto-Oncogene Proteins p21(ras)/metabolism , Pulmonary Alveoli/cytology , Respiratory Mucosa/cytology , Animals , DNA Primers/genetics , Flow Cytometry , In Situ Hybridization , Mice , Mice, Transgenic , Microarray Analysis , SOX9 Transcription Factor/metabolism , Xenopus laevis
7.
Nature ; 460(7251): 66-72, 2009 Jul 02.
Article in English | MEDLINE | ID: mdl-19571879

ABSTRACT

Stem cells are controlled, in part, by genetic pathways frequently dysregulated during human tumorigenesis. Either stimulation of Wnt/beta-catenin signalling or overexpression of telomerase is sufficient to activate quiescent epidermal stem cells in vivo, although the mechanisms by which telomerase exerts these effects are not understood. Here we show that telomerase directly modulates Wnt/beta-catenin signalling by serving as a cofactor in a beta-catenin transcriptional complex. The telomerase protein component TERT (telomerase reverse transcriptase) interacts with BRG1 (also called SMARCA4), a SWI/SNF-related chromatin remodelling protein, and activates Wnt-dependent reporters in cultured cells and in vivo. TERT serves an essential role in formation of the anterior-posterior axis in Xenopus laevis embryos, and this defect in Wnt signalling manifests as homeotic transformations in the vertebrae of Tert(-/-) mice. Chromatin immunoprecipitation of the endogenous TERT protein from mouse gastrointestinal tract shows that TERT physically occupies gene promoters of Wnt-dependent genes. These data reveal an unanticipated role for telomerase as a transcriptional modulator of the Wnt/beta-catenin signalling pathway.


Subject(s)
Chromatin/genetics , Signal Transduction , Telomerase/metabolism , Wnt Proteins/metabolism , Animals , Cell Line , Choristoma/genetics , Choristoma/pathology , DNA Helicases/metabolism , Genes, Reporter/genetics , HeLa Cells , Humans , Intestine, Small/metabolism , Mice , Nuclear Proteins/metabolism , Oocytes/cytology , Oocytes/growth & development , Plasmids/genetics , Promoter Regions, Genetic/genetics , Somites/abnormalities , Somites/embryology , Transcription Factors/metabolism , Wnt Proteins/genetics , Wnt3 Protein , Xenopus laevis/embryology , beta Catenin/genetics
8.
J Cell Sci ; 125(Pt 22): 5288-301, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22946057

ABSTRACT

A role for Rac1 GTPase in canonical Wnt signaling has recently been demonstrated, showing that it is required for ß-catenin translocation to the nucleus. In this study, we investigated the mechanism of Rac1 stimulation by Wnt. Upregulation of Rac1 activity by Wnt3a temporally correlated with enhanced p120-catenin binding to Rac1 and Vav2. Vav2 and Rac1 association with p120-catenin was modulated by phosphorylation of this protein, which was stimulated upon serine/threonine phosphorylation by CK1 and inhibited by tyrosine phosphorylation by Src or Fyn. Acting on these two post-translational modifications, Wnt3a induced the release of p120-catenin from E-cadherin, enabled the interaction of p120-catenin with Vav2 and Rac1, and facilitated Rac1 activation by Vav2. Given that p120-catenin depletion disrupts gastrulation in Xenopus, we analyzed p120-catenin mutants for their ability to rescue this phenotype. In contrast to the wild-type protein or other controls, p120-catenin point mutants that were deficient in the release from E-cadherin or in Vav2 or Rac1 binding failed to rescue p120-catenin depletion. Collectively, these results indicate that binding of p120-catenin to Vav2 and Rac1 is required for the activation of this GTPase upon Wnt signaling.


Subject(s)
Catenins/metabolism , Proto-Oncogene Proteins c-vav/metabolism , Wnt3A Protein/pharmacology , rac1 GTP-Binding Protein/metabolism , Animals , Cadherins/metabolism , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytosol/drug effects , Cytosol/metabolism , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Enzyme Activation/drug effects , Gastrulation/drug effects , Humans , Models, Biological , Mutant Proteins/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Phosphotyrosine/metabolism , Protein Binding/drug effects , Protein Transport/drug effects , Signal Transduction/drug effects , Xenopus/embryology , Xenopus/metabolism , beta Catenin/metabolism , Delta Catenin
9.
J Cell Sci ; 125(Pt 3): 561-9, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22389395

ABSTRACT

The Wnt pathways contribute to many processes in cancer and development, with ß-catenin being a key canonical component. p120-catenin, which is structurally similar to ß-catenin, regulates the expression of certain Wnt target genes, relieving repression conferred by the POZ- and zinc-finger-domain-containing transcription factor Kaiso. We have identified the kinase Dyrk1A as a component of the p120-catenin-Kaiso trajectory of the Wnt pathway. Using rescue and other approaches in Xenopus laevis embryos and mammalian cells, we found that Dyrk1A positively and selectively modulates p120-catenin protein levels, thus having an impact on p120-catenin and Kaiso (and canonical Wnt) gene targets such as siamois and wnt11. The Dyrk1A gene resides within the Down's syndrome critical region, which is amplified in Down's syndrome. A consensus Dyrk phosphorylation site in p120-catenin was identified, with a mutant mimicking phosphorylation exhibiting the predicted enhanced capacity to promote endogenous Wnt-11 and Siamois expression, and gastrulation defects. In summary, we report the biochemical and functional relationship of Dyrk1A with the p120-catenin-Kaiso signaling trajectory, with a linkage to canonical Wnt target genes. Conceivably, this work might also prove relevant to understanding the contribution of Dyrk1A dosage imbalance in Down's syndrome.


Subject(s)
Catenins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Repressor Proteins/metabolism , Wnt Signaling Pathway/physiology , Xenopus Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Catenins/genetics , DNA Primers/genetics , Down Syndrome/genetics , Down Syndrome/metabolism , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Molecular Sequence Data , Mutant Proteins/genetics , Mutant Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , RNA, Small Interfering/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Transfection , Xenopus Proteins/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics , Xenopus laevis/metabolism , Delta Catenin
10.
Nat Rev Cancer ; 5(12): 956-64, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16294216

ABSTRACT

Kaiso belongs to the zinc finger and broad-complex, tramtrack and bric-a-brac/poxvirus and zinc finger (BTB/POZ) protein family that has been implicated in tumorigenesis. Kaiso was first discovered in a complex with the armadillo-domain protein p120ctn and later shown to function as a transcriptional repressor. As p120ctn seems to relieve Kaiso-mediated repression, its altered intracellular localization in some cancer cells might result in aberrant Kaiso nuclear activity. Intriguingly, Kaiso's target genes include both methylated and sequence-specific recognition sites. The latter include genes that are modulated by the canonical Wnt (beta-catenin-T-cell factor) signalling pathway. Further interest in Kaiso stems from findings that its cytoplasmic versus nuclear localization is modulated by complex cues from the microenvironment.


Subject(s)
Neoplasms/genetics , Transcription Factors/genetics , Animals , Armadillo Domain Proteins/genetics , Catenins , Cell Adhesion Molecules , Cytoskeletal Proteins/genetics , Humans , Phosphoproteins , Repressor Proteins/genetics , Xenopus , Xenopus Proteins/genetics , beta Catenin/genetics , Delta Catenin
11.
Front Cell Neurosci ; 18: 1315941, 2024.
Article in English | MEDLINE | ID: mdl-38414752

ABSTRACT

Neuronal connectivity is regulated during normal brain development with the arrangement of spines and synapses being dependent on the morphology of dendrites. Further, in multiple neurodevelopmental and aging disorders, disruptions of dendrite formation or shaping is associated with atypical neuronal connectivity. We showed previously that Pdlim5 binds delta-catenin and promotes dendrite branching. We report here that Pdlim5 interacts with PalmD, a protein previously suggested by others to interact with the cytoskeleton (e.g., via adducin/spectrin) and to regulate membrane shaping. Functionally, the knockdown of PalmD or Pdlim5 in rat primary hippocampal neurons dramatically reduces branching and conversely, PalmD exogenous expression promotes dendrite branching as does Pdlim5. Further, we show that each proteins' effects are dependent on the presence of the other. In summary, using primary rat hippocampal neurons we reveal the contributions of a novel Pdlim5:PalmD protein complex, composed of functionally inter-dependent components responsible for shaping neuronal dendrites.

13.
bioRxiv ; 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37662414

ABSTRACT

Neuronal connectivity is regulated during normal brain development with the arrangement of spines and synapses being dependent on the morphology of dendrites. Further, in multiple neurodevelopmental and aging disorders, disruptions of dendrite formation or shaping is associated with atypical neuronal connectivity. We showed previously that Pdlim5 binds delta-catenin and promotes dendrite branching (Baumert et al., J Cell Biol 2020). We report here that Pdlim5 interacts with PalmD, a protein previously suggested by others to interact with the cytoskeleton (e.g., via adducin/ spectrin) and to regulate membrane shaping. Functionally, the knockdown of PalmD or Pdlim5 in rat primary hippocampal neurons dramatically reduces branching and conversely, PalmD exogenous expression promotes dendrite branching as does Pdlim5. Further, we show that effects of each protein are dependent on the presence of the other. In summary, using primary rat hippocampal neurons we reveal the contributions of a novel Pdlim5:PalmD protein complex, composed of functionally inter-dependent components responsible for shaping neuronal dendrites.

14.
Front Cell Neurosci ; 17: 1151249, 2023.
Article in English | MEDLINE | ID: mdl-37082208

ABSTRACT

Dendritic arborization is essential for proper neuronal connectivity and function. Conversely, abnormal dendrite morphology is associated with several neurological pathologies like Alzheimer's disease and schizophrenia. Among major intrinsic mechanisms that determine the extent of the dendritic arbor is cytoskeletal remodeling. Here, we characterize and compare the impact of the four proteins involved in cytoskeletal remodeling-vertebrate members of the p120-catenin subfamily-on neuronal dendrite morphology. In relation to each of their own distributions, we find that p120-catenin and delta-catenin are expressed at relatively higher proportions in growth cones compared to ARVCF-catenin and p0071-catenin; ARVCF-catenin is expressed at relatively high proportions in the nucleus; and all catenins are expressed in dendritic processes and the soma. Through altering the expression of each p120-subfamily catenin in neurons, we find that exogenous expression of either p120-catenin or delta-catenin correlates with increased dendritic length and branching, whereas their respective depletion decreases dendritic length and branching. While increasing ARVCF-catenin expression also increases dendritic length and branching, decreasing expression has no grossly observable morphological effect. Finally, increasing p0071-catenin expression increases dendritic branching, but not length, while decreasing expression decreases dendritic length and branching. These distinct localization patterns and morphological effects during neuron development suggest that these catenins have both shared and distinct roles in the context of dendrite morphogenesis.

15.
bioRxiv ; 2023 Oct 26.
Article in English | MEDLINE | ID: mdl-37961492

ABSTRACT

Aging is an inevitable process with senescence being one of its hallmarks. Recent advances have indicated that the elimination of senescent cells can reduce the signs of aging and increase healthy life span. Here, we identify a negative modulator of aging, Sprr1a, and in turn a negative modulator of Sprr1a, miR-130b. We show that reductions in Sprr1a levels, including via miR-130b expression, promotes cell senescence-like phenotype. We find that mediators of senescence, such as inflammatory cytokines and cell cycle regulators, are modulated by the miR-130b and Sprr1a-related pathway. For example, the levels of p16, p53 and p21 become decreased or increased upon the respective expression of Sprr1a versus miR-130b. Further, as shown in relation to p16 levels and ß-galactosidase levels, cells expressing Sprr1a exhibit significant protection from senescence-inducing factors such as radiation or Doxorubicin, suggesting that Sprr1a might contribute to protection against age-related pathologies. Taken together, we introduce two modulators of properties associated with senescence-like phenotype.

16.
J Biol Chem ; 286(26): 23178-88, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21561870

ABSTRACT

δ-Catenin is an Armadillo protein of the p120-catenin subfamily capable of modulating cadherin stability, small GTPase activity, and nuclear transcription. From yeast two-hybrid screening of a human embryonic stem cell cDNA library, we identified δ-catenin as a potential interacting partner of the caspase-3 protease, which plays essential roles in apoptotic as well as non-apoptotic processes. Interaction of δ-catenin with caspase-3 was confirmed using cleavage assays conducted in vitro, in Xenopus apoptotic extracts, and in cell line chemically induced contexts. The cleavage site, a highly conserved caspase consensus motif (DELD) within Armadillo repeat 6 of δ-catenin, was identified through peptide sequencing. Cleavage thus generates an amino-terminal (residues 1-816) and carboxyl-terminal (residues 817-1314) fragment, each containing about half of the central Armadillo domain. We found that cleavage of δ-catenin both abolishes its association with cadherins and impairs its ability to modulate small GTPases. Interestingly, 817-1314 possesses a conserved putative nuclear localization signal that may facilitate the nuclear targeting of δ-catenin in defined contexts. To probe for novel nuclear roles of δ-catenin, we performed yeast two-hybrid screening of a mouse brain cDNA library, resolving and then validating interaction with an uncharacterized KRAB family zinc finger protein, ZIFCAT. Our results indicate that ZIFCAT is nuclear and suggest that it may associate with DNA as a transcriptional repressor. We further determined that other p120 subfamily catenins are similarly cleaved by caspase-3 and likewise bind ZIFCAT. Our findings potentially reveal a simple yet novel signaling pathway based upon caspase-3 cleavage of p120-catenin subfamily members, facilitating the coordinate modulation of cadherins, small GTPases, and nuclear functions.


Subject(s)
Apoptosis/physiology , Caspase 3/metabolism , Catenins/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Amino Acid Motifs , Animals , Cadherins/genetics , Cadherins/metabolism , Caspase 3/genetics , Catenins/genetics , DNA-Binding Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Mice , Nuclear Proteins/genetics , Protein Structure, Tertiary , Repressor Proteins/genetics , Saccharomyces cerevisiae , Two-Hybrid System Techniques , Xenopus laevis , Delta Catenin
17.
J Cell Sci ; 123(Pt 24): 4351-65, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21098636

ABSTRACT

Wnt signaling pathways have fundamental roles in animal development and tumor progression. Here, employing Xenopus embryos and mammalian cell lines, we report that the degradation machinery of the canonical Wnt pathway modulates p120-catenin protein stability through mechanisms shared with those regulating ß-catenin. For example, in common with ß-catenin, exogenous expression of destruction complex components, such as GSK3ß and axin, promotes degradation of p120-catenin. Again in parallel with ß-catenin, reduction of canonical Wnt signals upon depletion of LRP5 and LRP6 results in p120-catenin degradation. At the primary sequence level, we resolved conserved GSK3ß phosphorylation sites in the amino-terminal region of p120-catenin present exclusively in isoform-1. Point-mutagenesis of these residues inhibited the association of destruction complex components, such as those involved in ubiquitylation, resulting in stabilization of p120-catenin. Functionally, in line with predictions, p120 stabilization increased its signaling activity in the context of the p120-Kaiso pathway. Importantly, we found that two additional p120-catenin family members, ARVCF-catenin and δ-catenin, associate with axin and are degraded in its presence. Thus, as supported using gain- and loss-of-function approaches in embryo and cell line systems, canonical Wnt signals appear poised to have an impact upon a breadth of catenin biology in vertebrate development and, possibly, human cancers.


Subject(s)
Catenins/metabolism , Signal Transduction , Wnt Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Axin Protein , Casein Kinase I/metabolism , Catenins/chemistry , Cell Line , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Molecular Sequence Data , Mutant Proteins/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Interaction Mapping , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Processing, Post-Translational , Protein Stability , Protein Structure, Tertiary , Repressor Proteins/metabolism , Ubiquitin/metabolism , Ubiquitination , Xenopus , Xenopus Proteins , Delta Catenin
18.
J Cell Sci ; 123(Pt 23): 4128-44, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21062899

ABSTRACT

In common with other p120-catenin subfamily members, Xenopus ARVCF (xARVCF) binds cadherin cytoplasmic domains to enhance cadherin metabolic stability or, when dissociated, modulates Rho-family GTPases. We report here that xARVCF binds and is stabilized by Xenopus KazrinA (xKazrinA), a widely expressed conserved protein that bears little homology to established protein families, and which is known to influence keratinocyte proliferation and differentiation and cytoskeletal activity. Although we found that xKazrinA binds directly to xARVCF, we did not resolve xKazrinA within a larger ternary complex with cadherin, nor did it co-precipitate with core desmosomal components. Instead, screening revealed that xKazrinA binds spectrin, suggesting a potential means by which xKazrinA localizes to cell-cell borders. This was supported by the resolution of a ternary biochemical complex of xARVCF-xKazrinA-xß2-spectrin and, in vivo, by the finding that ectodermal shedding followed depletion of xKazrin in Xenopus embryos, a phenotype partially rescued with exogenous xARVCF. Cell shedding appeared to be the consequence of RhoA activation, and thereby altered actin organization and cadherin function. Indeed, we also revealed that xKazrinA binds p190B RhoGAP, which was likewise capable of rescuing Kazrin depletion. Finally, xKazrinA was found to associate with δ-catenins and p0071-catenins but not with p120-catenin, suggesting that Kazrin interacts selectively with additional members of the p120-catenin subfamily. Taken together, our study supports the essential role of Kazrin in development, and reveals the biochemical and functional association of KazrinA with ARVCF-catenin, spectrin and p190B RhoGAP.


Subject(s)
Armadillo Domain Proteins/metabolism , Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Epithelial Cells/metabolism , GTPase-Activating Proteins/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Spectrin/metabolism , Xenopus Proteins/metabolism , Xenopus/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Armadillo Domain Proteins/chemistry , Armadillo Domain Proteins/genetics , Cadherins/genetics , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Cell Line , Epithelial Cells/chemistry , Epithelial Cells/enzymology , GTPase-Activating Proteins/genetics , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Phosphoproteins/chemistry , Phosphoproteins/genetics , Protein Binding , Protein Structure, Tertiary , Spectrin/genetics , Two-Hybrid System Techniques , Xenopus/genetics , Xenopus Proteins/chemistry , Xenopus Proteins/genetics , rhoA GTP-Binding Protein/genetics
19.
J Am Soc Nephrol ; 22(9): 1654-64, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21804089

ABSTRACT

Canonical ß-catenin-mediated Wnt signaling is essential for the induction of nephron development. Noncanonical Wnt/planar cell polarity (PCP) pathways contribute to processes such as cell polarization and cytoskeletal modulation in several tissues. Although PCP components likely establish the plane of polarization in kidney tubulogenesis, whether PCP effectors directly modulate the actin cytoskeleton in tubulogenesis is unknown. Here, we investigated the roles of Wnt PCP components in cytoskeletal assembly during kidney tubule morphogenesis in Xenopus laevis and zebrafish. We found that during tubulogenesis, the developing pronephric anlagen expresses Daam1 and its interacting Rho-GEF (WGEF), which compose one PCP/noncanonical Wnt pathway branch. Knockdown of Daam1 resulted in reduced expression of late pronephric epithelial markers with no apparent effect upon early markers of patterning and determination. Inhibiting various points in the Daam1 signaling pathway significantly reduced pronephric tubulogenesis. These data indicate that pronephric tubulogenesis requires the Daam1/WGEF/Rho PCP pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Polarity , Cytoskeleton/metabolism , Kidney Tubules/embryology , Organogenesis , Wnt Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Apoptosis , Cell Proliferation , Female , Guanine Nucleotide Exchange Factors/metabolism , Xenopus laevis , Zebrafish , Zebrafish Proteins/metabolism
20.
Dev Dyn ; 240(12): 2601-12, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22028074

ABSTRACT

The novel adaptor protein Kazrin associates with multifunctional entities including p120-subfamily members (ARVCF-, delta-, and p0071-catenin). Critical contributions of Kazrin to development or homeostasis are indicated with respect to ectoderm formation, integrity and keratinocyte differentiation, whereas its presence in varied tissues suggests broader roles. We find that Kazrin is maternally loaded, is expressed across development and becomes enriched in the forming head. Kazrin's potential contributions to craniofacial development were probed by means of knockdown in the prospective anterior neural region. Cartilaginous head structures as well as eyes on injected sides were reduced in size, with molecular markers suggesting an impact upon neural crest cell establishment and migration. Similar effects followed the depletion of ARVCF (or delta-catenin), with Kazrin:ARVCF functional interplay supported upon ARVCF partial rescue of Kazrin knockdown phenotypes. Thus, Kazrin and its associating ARVCF- and delta-catenins, are required to form craniofacial tissues originating from cranial neural crest and precordal plate.


Subject(s)
Armadillo Domain Proteins/metabolism , Catenins/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement/physiology , Membrane Proteins/metabolism , Neural Crest/embryology , Organogenesis/physiology , Phosphoproteins/metabolism , Skull/embryology , Xenopus Proteins/metabolism , Animals , Armadillo Domain Proteins/genetics , Cartilage/embryology , Catenins/genetics , Cell Adhesion Molecules/genetics , Eye/embryology , Gene Knockdown Techniques , Membrane Proteins/genetics , Phosphoproteins/genetics , Skull/metabolism , Xenopus Proteins/genetics , Xenopus laevis , Delta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL