Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 131(6)2021 03 15.
Article in English | MEDLINE | ID: mdl-33497368

ABSTRACT

Familial exudative vitreoretinopathy (FEVR) is a severe retinal vascular disease that causes blindness. FEVR has been linked to mutations in several genes associated with inactivation of the Norrin/ß-catenin signaling pathway, but these account for only approximately 50% of cases. We report that mutations in α-catenin (CTNNA1) cause FEVR by overactivating the ß-catenin pathway and disrupting cell adherens junctions. We identified 3 heterozygous mutations in CTNNA1 (p.F72S, p.R376Cfs*27, and p.P893L) by exome sequencing and further demonstrated that FEVR-associated mutations led to overactivation of Norrin/ß-catenin signaling as a result of impaired protein interactions within the cadherin-catenin complex. The clinical features of FEVR were reproduced in mice lacking Ctnna1 in vascular endothelial cells (ECs) or with overactivated ß-catenin signaling by an EC-specific gain-of-function allele of Ctnnb1. In isolated mouse lung ECs, both CTNNA1-P893L and F72S mutants failed to rescue either the disrupted F-actin arrangement or the VE-cadherin and CTNNB1 distribution. Moreover, we discovered that compound heterozygous Ctnna1 F72S and a deletion allele could cause a similar phenotype. Furthermore, in a FEVR family, we identified a mutation of LRP5, which activates Norrin/ß-catenin signaling, and the corresponding knockin mice exhibited a partial FEVR-like phenotype. Our study demonstrates that the precise regulation of ß-catenin activation is critical for retinal vascular development and provides new insights into the pathogenesis of FEVR.


Subject(s)
Eye Proteins/metabolism , Familial Exudative Vitreoretinopathies/genetics , Familial Exudative Vitreoretinopathies/metabolism , Nerve Tissue Proteins/metabolism , alpha Catenin/genetics , beta Catenin/metabolism , Amino Acid Sequence , Animals , Blood-Brain Barrier/metabolism , Disease Models, Animal , Familial Exudative Vitreoretinopathies/etiology , Female , Heterozygote , Humans , Male , Mice , Mice, Knockout , Mutation , Pedigree , Phenotype , Retinal Vessels/metabolism , Retinal Vessels/pathology , Signal Transduction/genetics , Exome Sequencing , alpha Catenin/deficiency , alpha Catenin/metabolism , beta Catenin/genetics
2.
Sci Rep ; 8(1): 1151, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29348601

ABSTRACT

Many cell lines derived from solid cancers can form spheroids, which recapitulate tumor cell clusters and are more representative of the in vivo situation than 2D cultures. During spheroid formation, a small proportion of a variety of different colon cancer cell lines did not integrate into the sphere and lost cell-cell adhesion properties. An enrichment protocol was developed to augment the proportion of these cells to 100% purity. The basis for the separation of spheroids from non-spheroid forming (NSF) cells is simple gravity-sedimentation. This protocol gives rise to sub-populations of colon cancer cells with stable loss of cell-cell adhesion. SW620 cells lacked E-cadherin, DLD-1 cells lost α-catenin and HCT116 cells lacked P-cadherin in the NSF state. Knockdown of these molecules in the corresponding spheroid-forming cells demonstrated that loss of the respective proteins were indeed responsible for the NSF phenotypes. Loss of the spheroid forming phenotype was associated with increased migration and invasion properties in all cell lines tested. Hence, we identified critical molecules involved in spheroid formation in different cancer cell lines. We present here a simple, powerful and broadly applicable method to generate new sublines of tumor cell lines to study loss of cell-cell adhesion in cancer progression.


Subject(s)
Antigens, CD/genetics , Cadherins/genetics , Cell Adhesion/genetics , Gene Expression Regulation, Neoplastic , Spheroids, Cellular/metabolism , alpha Catenin/genetics , Actins/genetics , Actins/metabolism , Cadherins/deficiency , Cell Communication , Cell Line, Tumor , Cell Movement , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/metabolism , HCT116 Cells , Humans , Karyotyping , Phenotype , Signal Transduction , Spheroids, Cellular/pathology , alpha Catenin/deficiency
3.
Sci Rep ; 4: 6835, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25355493

ABSTRACT

The liver is unique in its capacity to regenerate after injury, during which hepatocytes actively divide and establish cell-cell contacts through cell adhesion complexes. Here, we demonstrate that the loss of α-catenin, a well-established adhesion component, dramatically disrupts liver regeneration. Using a partial hepatectomy model, we show that regenerated livers from α-catenin knockdown mice are grossly larger than control regenerated livers, with an increase in cell size and proliferation. This increased proliferation correlated with increased YAP activation, implicating α-catenin in the Hippo/YAP pathway. Additionally, α-catenin knockdown mice exhibited a phenotype reminiscent of clinical cholestasis, with drastically altered bile canaliculi, elevated levels of bile components and signs of jaundice and inflammation. The disrupted regenerative capacity is a result of actin cytoskeletal disorganisation, leading to a loss of apical microvilli, dilated lumens in the bile canaliculi, and leaky tight junctions. This study illuminates a novel, essential role for α-catenin in liver regeneration.


Subject(s)
Cholestasis/genetics , Liver Regeneration/physiology , alpha Catenin/genetics , Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bile Canaliculi/pathology , Bile Canaliculi/ultrastructure , Cell Cycle Proteins , Cell Proliferation , Cholestasis/blood , Female , Hepatocytes/physiology , Mice , Mice, Knockout , Microvilli/ultrastructure , Models, Animal , Phosphoproteins/metabolism , YAP-Signaling Proteins , alpha Catenin/deficiency
4.
J Cell Sci ; 125(Pt 4): 1058-67, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22421363

ABSTRACT

It is generally accepted that the intercalated disc (ICD) required for mechano-electrical coupling in the heart consists of three distinct junctional complexes: adherens junctions, desmosomes and gap junctions. However, recent morphological and molecular data indicate a mixing of adherens junctional and desmosomal components, resulting in a 'hybrid adhering junction' or 'area composita'. The α-catenin family member αT-catenin, part of the N-cadherin-catenin adhesion complex in the heart, is the only α-catenin that interacts with the desmosomal protein plakophilin-2 (PKP2). Thus, it has been postulated that αT-catenin might serve as a molecular integrator of the two adhesion complexes in the area composita. To investigate the role of αT-catenin in the heart, gene targeting technology was used to delete the Ctnna3 gene, encoding αT-catenin, in the mouse. The αT-catenin-null mice are viable and fertile; however, the animals exhibit progressive cardiomyopathy. Adherens junctional and desmosomal proteins were unaffected by loss of αT-catenin, with the exception of the desmosomal protein PKP2. Immunogold labeling at the ICD demonstrated in the αT-catenin-null heart a preferential reduction of PKP2 at the area composita compared with the desmosome. Furthermore, gap junction protein Cx43 was reduced at the ICD, including its colocalization with N-cadherin. Gap junction remodeling in αT-catenin-knockout hearts was associated with an increased incidence of ventricular arrhythmias after acute ischemia. This novel animal model demonstrates for the first time how perturbation in αT-catenin can affect both PKP2 and Cx43 and thereby highlights the importance of understanding the crosstalk between the junctional proteins of the ICD and its implications for arrhythmogenic cardiomyopathy.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Cardiomyopathy, Dilated/pathology , Gap Junctions/metabolism , Heart Ventricles/physiopathology , Myocardial Ischemia/complications , Myocytes, Cardiac/metabolism , alpha Catenin/deficiency , Adherens Junctions/metabolism , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/pathology , Cadherins/metabolism , Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Dilated/physiopathology , Connexin 43/deficiency , Connexin 43/metabolism , Desmosomes/metabolism , Disease Models, Animal , Electrocardiography , Gap Junctions/pathology , Heart Ventricles/pathology , Mice , Mice, Knockout , Mutation , Myocardial Reperfusion Injury , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/pathology , Plakophilins/deficiency , Plakophilins/metabolism , alpha Catenin/genetics , alpha Catenin/metabolism
5.
Proc Natl Acad Sci U S A ; 109(13): 4886-91, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22411810

ABSTRACT

Cadherin/catenin-based adhesions coordinate cellular growth, survival, migration, and differentiation within a tissue by mechanically anchoring cells to their neighbors. They also intersect with diverse signaling pathways in development and cancer. Although the adhesive functions of adherens junction proteins are well characterized, their contribution to other signaling pathways is less well understood. Here, we show that ablation of α-catenin in the epidermis selectively induces apoptosis in suprabasal differentiating keratinocytes while sparing basal cell progenitors. This protection from death is coupled to elevated focal adhesion signaling, faster migration, and an altered distribution of growth factor receptors. We show that simultaneous depletion of α-catenin and focal adhesion kinase or p21-activated kinase eliminates basal cell protection as well as the elevated migration and proliferation of cells. The increased dependency of cells upon matrix interactions for their survival when cell-cell adhesions are destabilized has important implications for cancer progression and metastasis.


Subject(s)
Apoptosis , Epidermal Cells , Homeostasis , Integrins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Signal Transduction , Adherens Junctions/metabolism , Animals , Cell Adhesion , Cell Movement , Cell Proliferation , Cytoskeleton/metabolism , Epidermis/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/metabolism , Keratinocytes/cytology , Keratinocytes/enzymology , Mice , Mice, Knockout , Morphogenesis , Phosphorylation , Receptors, Cell Surface/metabolism , alpha Catenin/deficiency , alpha Catenin/metabolism , p21-Activated Kinases/metabolism
6.
J Cell Sci ; 125(Pt 1): 233-45, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22266901

ABSTRACT

α-catenin associates the cadherin-catenin complex with the actin cytoskeleton. α-catenin binds to ß-catenin, which links it to the cadherin cytoplasmic tail, and F-actin, but also to a multitude of actin-associated proteins. These interactions suggest a highly complex cadherin-actin interface. Moreover, mammalian αE-catenin has been implicated in a cadherin-independent cytoplasmic function in Arp2/3-dependent actin regulation, and in cell signaling. The function and regulation of individual molecular interactions of α-catenin, in particular during development, are not well understood. We have generated mutations in Drosophila α-Catenin (α-Cat) to investigate α-Catenin function in this model, and to establish a setup for testing α-Catenin-related constructs in α-Cat-null mutant cells in vivo. Our analysis of α-Cat mutants in embryogenesis, imaginal discs and oogenesis reveals defects consistent with a loss of cadherin function. Compromising components of the Arp2/3 complex or its regulator SCAR ameliorate the α-Cat loss-of-function phenotype in embryos but not in ovaries, suggesting negative regulatory interactions between α-Catenin and the Arp2/3 complex in some tissues. We also show that the α-Cat mutant phenotype can be rescued by the expression of a DE-cadherin::α-Catenin fusion protein, which argues against an essential cytosolic, cadherin-independent role of Drosophila α-Catenin.


Subject(s)
Adherens Junctions/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Gene Deletion , alpha Catenin/genetics , alpha Catenin/metabolism , Actin-Related Protein 2-3 Complex/genetics , Actin-Related Protein 2-3 Complex/metabolism , Adherens Junctions/genetics , Animals , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Cadherins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Embryonic Development/genetics , Female , Head/growth & development , Imaginal Discs/metabolism , Larva/growth & development , Male , Mutagenesis , Oogenesis/genetics , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Phenotype , Spectrin/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Zygote/metabolism , alpha Catenin/deficiency
7.
Mol Biol Cell ; 22(22): 4247-55, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21937720

ABSTRACT

Cadherin-catenin interactions play an important role in cadherin-mediated adhesion. Here we present strong evidence that in the cadherin-catenin complex α-catenin contributes to the binding strength of another catenin, p120, to the same complex. Specifically, we found that a ß-catenin-uncoupled cadherin mutant interacts much more weakly with p120 than its full-size counterpart and that it is rapidly endocytosed from the surface of A-431 cells. We also showed that p120 overexpression stabilizes this mutant on the cell surface. Examination of the α-catenin-deficient MDA-MB-468 cells and their derivates in which α-catenin was reintroduced showed that α-catenin reinforces E-cadherin-p120 association. Finally, a cross-linking analysis of the cadherin-catenin complex indicated that a large loop located in the middle of the p120 arm-repeat domain is in close spatial vicinity to the amino-terminal VH1 domain of α-catenin. The six amino acid-long extension of this loop, caused by an alternative splicing, weakens p120 binding to cadherin. The data suggest that α-catenin-p120 contact within the cadherin-catenin complex can regulate cadherin trafficking.


Subject(s)
Cadherins/metabolism , Catenins/metabolism , alpha Catenin/metabolism , beta Catenin/metabolism , Catenins/biosynthesis , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Humans , Mutation , Protein Binding , Protein Transport , Signal Transduction , alpha Catenin/deficiency , beta Catenin/genetics , Delta Catenin
8.
J Cell Biol ; 194(4): 631-42, 2011 Aug 22.
Article in English | MEDLINE | ID: mdl-21844209

ABSTRACT

Desmosomes are cell-cell adhesion structures that integrate cytoskeletal networks. In addition to binding intermediate filaments, the desmosomal protein desmoplakin (DP) regulates microtubule reorganization in the epidermis. In this paper, we identify a specific subset of centrosomal proteins that are recruited to the cell cortex by DP upon epidermal differentiation. These include Lis1 and Ndel1, which are centrosomal proteins that regulate microtubule organization and anchoring in other cell types. This recruitment was mediated by a region of DP specific to a single isoform, DPI. Furthermore, we demonstrate that the epidermal-specific loss of Lis1 results in dramatic defects in microtubule reorganization. Lis1 ablation also causes desmosomal defects, characterized by decreased levels of desmosomal components, decreased attachment of keratin filaments, and increased turnover of desmosomal proteins at the cell cortex. This contributes to loss of epidermal barrier activity, resulting in completely penetrant perinatal lethality. This work reveals essential desmosome-associated components that control cortical microtubule organization and unexpected roles for centrosomal proteins in epidermal function.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Desmosomes/metabolism , Epidermis/metabolism , Keratinocytes/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase/deficiency , 1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Animals , Carrier Proteins/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Desmoplakins/deficiency , Desmoplakins/genetics , Epidermis/embryology , Fluorescent Antibody Technique , Mice , Mice, Knockout , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Permeability , Protein Transport , Recombinant Fusion Proteins/metabolism , Transfection , alpha Catenin/deficiency , alpha Catenin/genetics
9.
Breast Cancer Res Treat ; 122(1): 125-33, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19763817

ABSTRACT

Mutations of E-cadherin have been identified in half of lobular breast cancers and diffuse-type gastric cancers, two tumor subtypes with remarkably similar pathological appearances including small rounded cells with scant cytoplasm and a diffuse growth pattern. A causal role for E-cadherin gene mutations in the lobular breast cancer phenotype was recently demonstrated in E-cadherin knock-out mice. These observations suggested that another gene in the E-cadherin tumor suppressor pathway might be mutated in lobular breast cancers with wild-type E-cadherin genes. Here, we identified E-cadherin gene mutations exclusively in human breast cancer cell lines that grow with a rounded cell morphology. Using expression analyses and gene mutation analyses, we have identified four biallelic inactivating alpha-catenin mutations among 55 human breast cancer cell lines. All four alpha-catenin mutations predicted premature termination of the encoded proteins, and concordantly, none of the four mutant cell lines expressed alpha-catenin proteins. Importantly, three of the alpha-catenin mutant cell lines had the rounded cell morphology and all 14 cell lines with the rounded cell morphology had mutations of either E-cadherin or alpha-catenin. As anticipated, loss of alpha-catenin protein expression was associated with the lobular subtype in primary breast cancers. Together, our observations suggest that alpha-catenin may be a new tumor suppressor gene that operates in the E-cadherin tumor suppressor pathway.


Subject(s)
Breast Neoplasms/pathology , Carcinoma/pathology , Codon, Nonsense , Genes, Tumor Suppressor , Neoplasm Proteins/genetics , alpha Catenin/genetics , Alleles , Breast Neoplasms/genetics , Cadherins/genetics , Cadherins/physiology , Carcinoma/genetics , Carcinoma, Lobular/genetics , Carcinoma, Lobular/pathology , Cell Adhesion/genetics , Cell Line, Tumor/metabolism , Cell Shape/genetics , DNA Methylation , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/deficiency , Oligonucleotide Array Sequence Analysis , alpha Catenin/biosynthesis , alpha Catenin/deficiency
10.
J Cell Sci ; 121(Pt 9): 1357-62, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18397997

ABSTRACT

beta-Catenin is a crucial mediator of the canonical Wnt-signaling pathway. alpha-catenin is a major beta-catenin-binding protein, and overexpressed alpha-catenin can negatively regulate beta-catenin activity. Thus, alpha-catenin may be an important modulator of the Wnt pathway. We show here that endogenous alpha-catenin has little impact on the transcriptional activity of beta-catenin in developing mammalian organisms. We analyzed beta-catenin signaling in mice with conditional deletion of alphaE-catenin (Ctnna1) in the developing central nervous system. This mutation results in brain hyperplasia and we investigated whether activation of beta-catenin signaling may be at least partially responsible for this phenotype. To reveal potential quantitative or spatial changes in beta-catenin signaling, we used mice carrying a beta-catenin-signaling reporter transgene. In addition, we analyzed the expression of known endogenous targets of the beta-catenin pathway and the amount and localization of beta-catenin in mutant progenitor cells. We found that although loss of alphaE-catenin resulted in disruption of intercellular adhesion and hyperplasia in the developing brain, beta-catenin signaling was not altered. We conclude that endogenous alphaE-catenin has no significant impact on beta-catenin transcriptional activities in the developing mammalian brain.


Subject(s)
Brain/embryology , Mammals/embryology , Signal Transduction , alpha Catenin/metabolism , beta Catenin/metabolism , Animals , Brain/pathology , Cell Nucleus/metabolism , Genes, Reporter , Hyperplasia , Protein Binding , Protein Transport , TCF Transcription Factors/metabolism , Transcriptional Activation/genetics , alpha Catenin/deficiency , beta Catenin/genetics
11.
Circulation ; 114(10): 1046-55, 2006 Sep 05.
Article in English | MEDLINE | ID: mdl-16923756

ABSTRACT

BACKGROUND: alpha-E-catenin is a cell adhesion protein, located within the adherens junction, thought to be essential in directly linking the cadherin-based adhesion complex to the actin cytoskeleton. Although alpha-E-catenin is expressed in the adherens junction of the cardiomyocyte intercalated disc, and perturbations in its expression are observed in models of dilated cardiomyopathy, its role in the myocardium remains unknown. METHODS AND RESULTS: To determine the effects of alpha-E-catenin on cardiomyocyte ultrastructure and disease, we generated cardiac-specific alpha-E-catenin conditional knockout mice (alpha-E-cat cKO). alpha-E-cat cKO mice displayed progressive dilated cardiomyopathy and unique defects in the right ventricle. The effects on cardiac morphology/function in alpha-E-cat cKO mice were preceded by ultrastructural defects in the intercalated disc and complete loss of vinculin at the intercalated disc. alpha-E-cat cKO mice also revealed a striking susceptibility of the ventricular free wall to rupture after myocardial infarction. CONCLUSIONS: These results demonstrate a clear functional role for alpha-E-catenin in the cadherin/catenin/vinculin complex in the myocardium in vivo. Ablation of alpha-E-catenin within this complex leads to defects in cardiomyocyte structural integrity that result in unique forms of cardiomyopathy and predisposed susceptibility to death after myocardial stress. These studies further highlight the importance of studying the role of alpha-E-catenin in human cardiac injury and cardiomyopathy in the future.


Subject(s)
Cardiomyopathy, Dilated/genetics , Muscle Cells/physiology , Myocardial Infarction/genetics , Ventricular Dysfunction, Right/genetics , alpha Catenin/genetics , Animals , Disease Models, Animal , Electrocardiography , Mice , Mice, Knockout , Muscle Cells/pathology , Myocardium/pathology , Proteins/metabolism , alpha Catenin/deficiency
12.
Dev Dyn ; 235(9): 2559-66, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16691566

ABSTRACT

Alpha N-catenin is a cadherin-binding protein, widely expressed in the nervous system; and it plays a crucial role in cadherin-mediated cell-cell adhesion. Here we report the effects of alpha N-catenin gene deficiency on brain morphogenesis. In addition to the previously reported phenotypes, we found that some of the axon tracts did not normally develop, in particular, axons of the anterior commissure failed to cross the midline, migrating, rather, to ectopic places. In restricted nuclei, a population of neurons was missing or their laminar arrangement was distorted. The ventricular structures were also deformed. These results indicate that alpha N-catenin has diverse roles in the organization of the central nervous system, but only in limited portions of the brain.


Subject(s)
Brain/embryology , Brain/metabolism , Nerve Tissue Proteins/deficiency , alpha Catenin/deficiency , Animals , Axons/metabolism , Axons/ultrastructure , Cell Movement , Female , Gene Expression Regulation, Developmental , In Situ Hybridization , Mammillary Bodies/embryology , Mammillary Bodies/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Olivary Nucleus/embryology , Olivary Nucleus/metabolism , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Septal Nuclei/embryology , Septal Nuclei/metabolism , alpha Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...