Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Cell Signal ; 76: 109786, 2020 12.
Article in English | MEDLINE | ID: mdl-32966883

ABSTRACT

BACKGROUND: Salt-inducible kinase 2 (SIK2) is abundant in adipocytes, but downregulated in adipose tissue from individuals with obesity and insulin resistance. Moreover, SIK isoforms are required for normal insulin signalling and glucose uptake in adipocytes, but the underlying molecular mechanisms are currently not known. The adherens junction protein JUP, also termed plakoglobin or γ-catenin, has recently been reported to promote insulin signalling in muscle cells. OBJECTIVE: The objective of this study was to analyse if JUP is required for insulin signalling in adipocytes and the underlying molecular mechanisms of this regulation. METHODS: Co-expression of SIK2 and JUP mRNA levels in adipose tissue from a human cohort was analysed. siRNA silencing and/or pharmacological inhibition of SIK2, JUP, class IIa HDACs and CRTC2 was employed in 3T3-L1- and primary rat adipocytes. JUP protein expression was analysed by western blot and mRNA levels by qPCR. Insulin signalling was evaluated by western blot as levels of phosphorylated PKB/Akt and AS160, and by monitoring the uptake of 3H-2-deoxyglucose. RESULTS: mRNA expression of SIK2 correlated with that of JUP in human adipose tissue. SIK2 inhibition or silencing resulted in downregulation of JUP mRNA and protein expression in 3T3-L1- and in primary rat adipocytes. Moreover, JUP silencing reduced the expression of PKB and the downstream substrate AS160, and consequently attenuated activity in the insulin signalling pathway, including insulin-induced glucose uptake. The known SIK2 substrates CRTC2 and class IIa HDACs were found to play a role in the SIK-mediated regulation of JUP expression. CONCLUSIONS: These findings identify JUP as a novel player in the regulation of insulin sensitivity in adipocytes, and suggest that changes in JUP expression could contribute to the effect of SIK2 on insulin signalling in these cells.


Subject(s)
Adipocytes , Glucose/metabolism , Insulin Resistance , Obesity/metabolism , Protein Serine-Threonine Kinases/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adipocytes/pathology , Animals , Cohort Studies , Female , Humans , Mice , Primary Cell Culture , Rats , Rats, Sprague-Dawley , gamma Catenin/physiology
2.
Mol Biol Cell ; 31(8): 741-752, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32049581

ABSTRACT

Collective cell migration plays crucial roles in tissue remodeling, wound healing, and cancer cell invasion. However, its underlying mechanism remains unknown. Previously, we showed that the RhoA-targeting guanine nucleotide exchange factor Solo (ARHGEF40) is required for tensile force-induced RhoA activation and proper organization of keratin-8/keratin-18 (K8/K18) networks. Here, we demonstrate that Solo knockdown significantly increases the rate at which Madin-Darby canine kidney cells collectively migrate on collagen gels. However, it has no apparent effect on the migratory speed of solitary cultured cells. Therefore, Solo decelerates collective cell migration. Moreover, Solo localized to the anteroposterior regions of cell-cell contact sites in collectively migrating cells and was required for the local accumulation of K8/K18 filaments in the forward areas of the cells. Partial Rho-associated protein kinase (ROCK) inhibition or K18 or plakoglobin knockdown also increased collective cell migration velocity. These results suggest that Solo acts as a brake for collective cell migration by generating pullback force at cell-cell contact sites via the RhoA-ROCK pathway. It may also promote the formation of desmosomal cell-cell junctions related to K8/K18 filaments and plakoglobin.


Subject(s)
Cell Movement/physiology , Signal Transduction/physiology , rho GTP-Binding Proteins/physiology , rho-Associated Kinases/physiology , Amides/pharmacology , Animals , Cell Polarity , Collagen , Cytoskeleton/physiology , Desmosomes/physiology , Dogs , Gels , Gene Knockdown Techniques , Keratin-18/antagonists & inhibitors , Keratin-18/genetics , Keratin-18/physiology , Keratin-8/antagonists & inhibitors , Keratin-8/genetics , Keratin-8/physiology , Madin Darby Canine Kidney Cells , Pyridines/pharmacology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Stress, Mechanical , Time-Lapse Imaging , gamma Catenin/antagonists & inhibitors , gamma Catenin/genetics , gamma Catenin/physiology , rac1 GTP-Binding Protein/physiology , rho GTP-Binding Proteins/antagonists & inhibitors , rhoA GTP-Binding Protein/physiology
3.
Lab Invest ; 98(11): 1375-1383, 2018 11.
Article in English | MEDLINE | ID: mdl-29802338

ABSTRACT

We generated cornea-specific plakoglobin (Jup; junctional plakoglobin) knockout mice in order to investigate the function of plakoglobin on the maintenance of the homeostasis of corneal epithelium in mice. Cornea epithelium-specific conditional knockouts (JupCEΔ/CEΔ) (cKO) were obtained by breeding keratin12-Cre (Krt12-Cre) mice to Jup-floxed (Jupf/f) mice. Light and transmission electron microscopic and immunohistochemical analyses were carried out to determine consequence of the loss of plakoglobin on maintaining corneal epithelium integrity under mechanical stress, e.g., brushing and wound healing. Immunohistochemistry analysis demonstrated that, although Jup ablation did not affect BrdU incorporation, basal cell-like cells labeled for keratin 14 were ectopically present in the supra-basal layer in mutant corneal epithelium, suggestive of altered cell differentiation. Plakoglobin-deficient epithelium exhibits increased fragility against mechanical intervention when compared to wild-type controls under identical treatment. Closure of an epithelial defect was significantly delayed in JupCEΔ/CEΔ epithelium. Our findings indicate that the lack of plakoglobin significantly affects corneal epithelium differentiation, as well as its structural integrity. Plakoglobin is essential to the maintenance of the structure of the corneal epithelium and its wound healing.


Subject(s)
Epithelium, Corneal/physiology , Wound Healing , gamma Catenin/physiology , Animals , Corneal Injuries , Epithelium, Corneal/ultrastructure , Mice, Transgenic
4.
Hepatology ; 67(6): 2320-2337, 2018 06.
Article in English | MEDLINE | ID: mdl-29023813

ABSTRACT

ß-Catenin, the downstream effector of the Wnt signaling, plays important roles in hepatic development, regeneration, and tumorigenesis. However, its role at hepatocyte adherens junctions (AJ) is relatively poorly understood, chiefly due to spontaneous compensation by γ-catenin. We simultaneously ablated ß- and γ-catenin expression in mouse liver by interbreeding ß-catenin-γ-catenin double-floxed mice and Alb-Cre transgenic mice. Double knockout mice show failure to thrive, impaired hepatocyte differentiation, cholemia, ductular reaction, progressive cholestasis, inflammation, fibrosis, and tumorigenesis, which was associated with deregulation of tight junctions (TJ) and bile acid transporters, leading to early morbidity and mortality, a phenotype reminiscent of progressive familial intrahepatic cholestasis (PFIC). To address the mechanism, we specifically and temporally eliminated both catenins from hepatocytes using adeno-associated virus 8 carrying Cre-recombinase under the thyroid-binding globulin promoter (AAV8-TBG-Cre). This led to a time-dependent breach of the blood-biliary barrier associated with sequential disruption of AJ and TJ verified by ultrastructural imaging and intravital microscopy, which revealed unique paracellular leaks around individual hepatocytes, allowing mixing of blood and bile and leakage of blood from one sinusoid to another. Molecular analysis identified sequential losses of E-cadherin, occludin, claudin-3, and claudin-5 due to enhanced proteasomal degradation, and of claudin-2, a ß-catenin transcriptional target, which was also validated in vitro. CONCLUSION: We report partially redundant function of catenins at AJ in regulating TJ and contributing to the blood-biliary barrier. Furthermore, concomitant hepatic loss of ß- and γ-catenin disrupts structural and functional integrity of AJ and TJ via transcriptional and posttranslational mechanisms. Mice with dual catenin loss develop progressive intrahepatic cholestasis, providing a unique model to study diseases such as PFIC. (Hepatology 2018;67:2320-2337).


Subject(s)
Adherens Junctions , Cholestasis, Intrahepatic/etiology , Tight Junctions , beta Catenin/physiology , gamma Catenin/physiology , Animals , Female , Hepatocytes , Male , Mice , Mice, Knockout , beta Catenin/genetics , gamma Catenin/genetics
5.
Am J Pathol ; 185(12): 3274-89, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26485505

ABSTRACT

γ-Catenin, an important component of desmosomes, may also participate in Wnt signaling. Herein, we dissect the role of γ-catenin in liver by generating conditional γ-catenin knockout (KO) mice and assessing their phenotype after bile duct ligation (BDL) and diethylnitrosamine-induced chemical carcinogenesis. At baseline, KO and wild-type littermates showed comparable serum biochemistry, liver histology, and global gene expression. ß-Catenin protein was modestly increased without any change in Wnt signaling. Desmosomes were maintained in KO, and despite no noticeable changes in gene expression, differential detergent fractionation revealed quantitative and qualitative changes in desmosomal cadherins, plaque proteins, and ß-catenin. Enhanced association of ß-catenin to desmoglein-2 and plakophilin-3 was observed in KO. When subjected to BDL, wild-type littermates showed specific changes in desmosomal protein expression. In KO, BDL deteriorated baseline compensatory changes, which manifested as enhanced injury and fibrosis. KO also showed enhanced tumorigenesis to diethylnitrosamine treatment because of Wnt activation, as also verified in vitro. γ-Catenin overexpression in hepatoma cells increased its binding to T-cell factor 4 at the expense of ß-catenin-T-cell factor 4 association, induced unique target genes, affected Wnt targets, and reduced cell proliferation and viability. Thus, γ-catenin loss in liver is basally well tolerated. However, after insults like BDL, these compensations at desmosomes fail, and KO show enhanced injury. Also, γ-catenin negatively regulates tumor growth by affecting Wnt signaling.


Subject(s)
Cholestasis/metabolism , Desmosomes/metabolism , Liver Neoplasms, Experimental/metabolism , Liver/metabolism , gamma Catenin/physiology , Animals , Bile Ducts/surgery , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cholestasis/pathology , Diethylnitrosamine , Female , Gene Expression Regulation/physiology , Gene Expression Regulation, Neoplastic/physiology , Liver Neoplasms, Experimental/pathology , Male , Mice, Knockout , Neoplasm Proteins/metabolism , Neoplasm Proteins/physiology , Signal Transduction/physiology , Tumor Cells, Cultured , Wnt Signaling Pathway/physiology , beta Catenin/physiology , gamma Catenin/deficiency , gamma Catenin/genetics
6.
J Clin Invest ; 125(4): 1708-12, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25705887

ABSTRACT

Arrhythmogenic cardiomyopathy (AC) is associated with mutations in genes encoding intercalated disc proteins and ultimately results in sudden cardiac death. A subset of patients with AC have the autosomal recessive cardiocutaneous disorder Naxos disease, which is caused by a 2-base pair deletion in the plakoglobin-encoding gene JUP that results in a truncated protein with reduced expression. In mice, cardiomyocyte-specific plakoglobin deficiency recapitulates many aspects of human AC, and overexpression of the truncated Naxos-associated plakoglobin also results in an AC-like phenotype; therefore, it is unclear whether Naxos disease results from loss or gain of function consequent to the plakoglobin mutation. Here, we generated 2 knockin mouse models in which endogenous Jup was engineered to express the Naxos-associated form of plakoglobin. In one model, Naxos plakoglobin bypassed the nonsense-mediated mRNA decay pathway, resulting in normal levels of the truncated plakoglobin. Moreover, restoration of Naxos plakoglobin to WT levels resulted in normal heart function. Together, these data indicate that a gain of function in the truncated form of the protein does not underlie the clinical phenotype of patients with Naxos disease and instead suggest that insufficiency of the truncated Naxos plakoglobin accounts for disease manifestation. Moreover, these results suggest that increasing levels of truncated or WT plakoglobin has potential as a therapeutic approach to Naxos disease.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia/genetics , Desmoplakins/genetics , Hair Diseases/genetics , Keratoderma, Palmoplantar/genetics , Myocytes, Cardiac/pathology , gamma Catenin/physiology , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Arrhythmogenic Right Ventricular Dysplasia/pathology , Codon, Nonsense , Fibrosis , Frameshift Mutation , Gene Knock-In Techniques , Genes, Lethal , Hair Diseases/pathology , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Humans , Keratoderma, Palmoplantar/pathology , Mice , Myocardial Contraction , Myocardium/pathology , Myocytes, Cardiac/metabolism , Nonsense Mediated mRNA Decay , Peptide Fragments/physiology , Phenotype , RNA Stability , RNA, Messenger/metabolism , Sequence Deletion , Wnt Signaling Pathway , gamma Catenin/chemistry , gamma Catenin/deficiency , gamma Catenin/genetics
7.
J Invest Dermatol ; 133(12): 2732-2740, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23652796

ABSTRACT

Desmosomes are cell adhesion junctions required for the normal development and maintenance of mammalian tissues and organs such as the skin, skin appendages, and the heart. The goal of this study was to investigate how desmocollins (DSCs), transmembrane components of desmosomes, are regulated at the transcriptional level. We hypothesized that differential expression of the Dsc2 and Dsc3 genes is a prerequisite for normal development of skin appendages. We demonstrate that plakoglobin (Pg) in conjunction with lymphoid enhancer-binding factor 1 (Lef-1) differentially regulates the proximal promoters of these two genes. Specifically, we found that Lef-1 acts as a switch activating Dsc2 and repressing Dsc3 in the presence of Pg. Interestingly, we also determined that NF-κB pathway components, the downstream effectors of the ectodysplasin-A (EDA)/ ectodysplasin-A receptor (EDAR)/NF-κB signaling cascade, can activate Dsc2 expression. We hypothesize that Lef-1 and EDA/EDAR/NF-κB signaling contribute to a shift in Dsc isoform expression from Dsc3 to Dsc2 in placode keratinocytes. It is tempting to speculate that this shift is required for the invasive growth of placode keratinocytes into the dermis, a crucial step in skin appendage formation.


Subject(s)
Gene Expression Regulation , Membrane Glycoproteins/metabolism , gamma Catenin/physiology , Animals , Binding Sites , Cell Adhesion , Desmocollins , Dogs , Ectodysplasins/metabolism , Keratinocytes/cytology , Lymphoid Enhancer-Binding Factor 1/metabolism , Madin Darby Canine Kidney Cells , Mice , Mice, Transgenic , NF-kappa B/metabolism , Promoter Regions, Genetic , Receptors, Ectodysplasin/metabolism , Skin/metabolism
8.
PLoS One ; 7(7): e42132, 2012.
Article in English | MEDLINE | ID: mdl-22860065

ABSTRACT

Plakoglobin (PG) is an armadillo protein that associates with both classic and desmosomal cadherins, but is primarily concentrated in mature desmosomes in epithelia. While reduced levels of PG have been reported in localized and hormone refractory prostate tumors, the functional significance of these changes is unknown. Here we report that PG expression is reduced in samples of a prostate tumor tissue array and inversely correlated with advancing tumor potential in 7 PCa cell lines. Ectopically expressed PG enhanced intercellular adhesive strength, and attenuated the motility and invasion of aggressive cell lines, whereas silencing PG in less tumorigenic cells had the opposite effect. PG also regulated cell-substrate adhesion and motility through extracellular matrix (ECM)-dependent inhibition of Src kinase, suggesting that PG's effects were not due solely to increased intercellular adhesion. PG silencing resulted in elevated levels of the ECM protein vitronectin (VN), and exposing PG-expressing cells to VN induced Src activity. Furthermore, increased VN levels and Src activation correlated with diminished expression of PG in patient tissues. Thus, PG may inhibit Src by keeping VN low. Our results suggest that loss of intercellular adhesion due to reduced PG expression might be exacerbated by activation of Src through a PG-dependent mechanism. Furthermore, PG down-regulation during PCa progression could contribute to the known VN-dependent promotion of PCa invasion and metastasis, demonstrating a novel functional interaction between desmosomal cell-cell adhesion and cell-substrate adhesion signaling axes in prostate cancer.


Subject(s)
Cell Adhesion/physiology , Cell Movement/physiology , Oncogene Protein pp60(v-src)/metabolism , Prostatic Neoplasms/pathology , Signal Transduction/physiology , Vitronectin/metabolism , gamma Catenin/physiology , Base Sequence , Cell Line, Tumor , Desmosomes/metabolism , Humans , Male , RNA, Small Interfering , Tissue Array Analysis
9.
PLoS One ; 6(10): e27064, 2011.
Article in English | MEDLINE | ID: mdl-22046445

ABSTRACT

Control over cell viability is a fundamental property underlying numerous physiological processes. Cell spreading on a substrate was previously demonstrated to be a major factor in determining the viability of individual cells. In multicellular organisms, cell-cell contact is likely to play a significant role in regulating cell vitality, but its function is easily masked by cell-substrate interactions, thus remains incompletely characterized. In this study, we show that suspended immortalized human keratinocyte sheets with persisting intercellular contacts exhibited significant contraction, junctional actin localization, and reinforcement of cell-cell adhesion strength. Further, cells within these sheets remain viable, in contrast to trypsinized cells suspended without either cell-cell or cell-substrate contact, which underwent apoptosis at high rates. Suppression of plakoglobin weakened cell-cell adhesion in cell sheets and suppressed apoptosis in suspended, trypsinized cells. These results demonstrate that cell-cell contact may be a fundamental control mechanism governing cell viability and that the junctional protein plakoglobin is a key regulator of this process. Given the near-ubiquity of plakoglobin in multicellular organisms, these findings could have significant implications for understanding cell adhesion, modeling disease progression, developing therapeutics and improving the viability of tissue engineering protocols.


Subject(s)
Cell Adhesion/physiology , Cell Survival , Keratinocytes/cytology , gamma Catenin/physiology , Apoptosis , Cells, Cultured , Cytoskeletal Proteins , Humans
10.
Am J Pathol ; 177(6): 2921-37, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21075858

ABSTRACT

Desmoglein 1 (Dsg1) is a desmosomal cadherin that is essential to epidermal integrity. In the blistering diseases bullous impetigo and staphylococcal scalded-skin syndrome, pathogenesis depends on cleavage of Dsg1 by a bacterial protease, exfoliative toxin A, which removes residues 1 to 381 of the Dsg1 ectodomain. However, the cellular responses to Dsg1 cleavage that precipitate keratinocyte separation to induce blister formation are unknown. Here, we show that ectodomain-deleted Dsg1 (Δ381-Dsg1) mimics the toxin-cleaved cadherin, disrupts desmosomes, and reduces the mechanical integrity of keratinocyte sheets. In addition, we demonstrate that truncated Dsg1 remains associated with its catenin partner, plakoglobin, and causes a reduction in the levels of endogenous desmosomal cadherins in a dose-dependent manner, leading us to hypothesize that plakoglobin sequestration by truncated Dsg1 destabilizes other cadherins. Accordingly, a triple-point mutant of the ectodomain-deleted cadherin, which is uncoupled from plakoglobin, does not impair adhesion, indicating that this interaction is essential to the pathogenic potential of truncated Dsg1. Moreover, we demonstrate that increasing plakoglobin levels rescues cadherin expression, desmosome organization, and functional adhesion in cells expressing Δ381-Dsg1 or treated with exfoliative toxin A. Finally, we report that histone deacetylase inhibition up-regulates desmosomal cadherins and prevents the loss of adhesion induced by Dsg1 truncation. These findings further our understanding of the mechanism of exfoliative toxin-induced pathology and suggest novel strategies to suppress blistering in bulbous impetigo and staphylococcal scalded-skin syndrome.


Subject(s)
Blister/etiology , Dermatitis, Exfoliative/etiology , Desmoglein 1/genetics , Exfoliatins/adverse effects , Protein Interaction Domains and Motifs/genetics , Sequence Deletion/physiology , gamma Catenin/physiology , Blister/genetics , Blister/pathology , Cell Adhesion/genetics , Cells, Cultured , Dermatitis, Exfoliative/genetics , Dermatitis, Exfoliative/pathology , Desmoglein 1/chemistry , Desmoglein 1/metabolism , Desmoglein 1/physiology , Desmosomal Cadherins/chemistry , Desmosomal Cadherins/genetics , Desmosomal Cadherins/metabolism , Desmosomal Cadherins/physiology , Exfoliatins/pharmacology , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , Keratinocytes/physiology , Protein Interaction Domains and Motifs/physiology , Protein Processing, Post-Translational/drug effects , Skin/metabolism , Skin/pathology , Staphylococcal Skin Infections/genetics , Staphylococcal Skin Infections/metabolism , Staphylococcal Skin Infections/pathology , Transduction, Genetic , gamma Catenin/genetics , gamma Catenin/metabolism
12.
Exp Dermatol ; 18(8): 658-68, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19558499

ABSTRACT

Both skin and heart are subject to shear mechanical stress and need to be stress-resistant in a flexible way. The intercellular connecting structures in skin and heart, the desmosomes, that have to resist these forces show remarkable resemblance in epidermis and myocardium. Mutations in desmosomal proteins lead to inherited desmosomal cardiocutaneous syndromes (DCCS): une liaison dangereuse. This article will critically review the cutaneous and cardiac features as well as the molecular background of DCCS, such as Naxos disease and Carvajal syndrome caused by deficiencies of plakoglobin and desmoplakin respectively. In addition, potential other desmosomal gene candidates for an involvement in cardiocutaneous syndromes are considered. The skin features in these syndromes may be the hallmark for the presence of progressive and ultimately lethal cardiac disease. Knowledge of these skin features and early recognition of such a syndrome may provide opportunities to halt or slow down cardiac disease progression, treat arrhythmias and even prevent sudden death.


Subject(s)
Heart Diseases/physiopathology , Myocardium/pathology , Skin Diseases/physiopathology , Skin/pathology , Animals , Desmoplakins/physiology , Desmosomes/physiology , Disease Progression , Heart/anatomy & histology , Heart Diseases/diagnosis , Humans , Mice , Microscopy, Electron/methods , Mutation , Plectin/physiology , Skin/anatomy & histology , Skin Diseases/diagnosis , gamma Catenin/physiology
13.
Dev Biol ; 327(1): 83-96, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19101534

ABSTRACT

Plakoglobin, or gamma-catenin, is found in both desmosomes and adherens junctions and participates in Wnt signalling. Mutations in the human gene are implicated in the congenital heart disorder, arrhythmogenic right ventricular cardiomyopathy (ARVC), but the signalling effects of plakoglobin loss in ARVC have not been established. Here we report that knockdown of plakoglobin in zebrafish results in decreased heart size, reduced heartbeat, cardiac oedema, reflux of blood between heart chambers and a twisted tail. Wholemount in situ hybridisation shows reduced expression of the heart markers nkx2.5 at 24 hours post fertilisation (hpf), and cmlc2 and vmhc at 48 hpf, while there is lack of restriction of the valve markers notch1b and bmp4 at 48 hpf. Wnt target gene expression was examined by semi-quantitative RT-PCR and found to be increased in morphant embryos indicating that plakoglobin is antagonistic to Wnt signalling. Co-expression of the Wnt inhibitor, Dkk1, rescues the cardiac phenotype of the plakoglobin morphant. beta-catenin protein expression is increased in morphant embryos as is its colocalisation with E-cadherin in adherens junctions. Endothelial cells at the atrioventricular boundary of morphant hearts have an aberrant morphology, indicating problems with valvulogenesis. Morphants also have decreased numbers of desmosomes and adherens junctions in the intercalated discs. These results establish the zebrafish as a model for ARVC caused by loss of plakoglobin function and indicate that there are signalling as well as structural consequences of this loss.


Subject(s)
Heart/growth & development , Signal Transduction , Zebrafish/growth & development , gamma Catenin/physiology , Animals , Embryo, Nonmammalian , Endothelial Cells/pathology , Gene Expression Profiling , Heart/embryology , Heart Defects, Congenital/genetics , Intercellular Junctions , Phenotype , Wnt Proteins/metabolism , gamma Catenin/genetics
14.
Dev Biol ; 319(2): 298-308, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18514185

ABSTRACT

Tissue morphogenesis and maintenance of complex tissue architecture requires a variety of cell-cell junctions. Typically, cells adhere to one another through cadherin junctions, both adherens and desmosomal junctions, strengthened by association with cytoskeletal networks during development. Both beta- and gamma-catenins are reported to link classical cadherins to the actin cytoskeleton, but only gamma-catenin binds to the desmosomal cadherins, which links them to intermediate filaments through its association with desmoplakin. Here we provide the first biochemical evidence that, in vivo, gamma-catenin also mediates interactions between classical cadherins and the intermediate filament cytoskeleton, linked through desmoplakin. In the developing lens, which has no desmosomes, we discovered that vimentin became linked to N-cadherin complexes in a differentiation-state specific manner. This newly identified junctional complex was tissue specific but not unique to the lens. To determine whether in this junction N-cadherin was linked to vimentin through gamma-catenin or beta-catenin we developed an innovative "double" immunoprecipitation technique. This approach made possible, for the first time, the separation of N-cadherin/gamma-catenin from N-cadherin/beta-catenin complexes and the identification of multiple members of each of these isolated protein complexes. The study revealed that vimentin was associated exclusively with N-cadherin/gamma-catenin junctions. Assembly of this novel class of cadherin junctions was coincident with establishment of the unique cytoarchitecture of lens fiber cells. In addition, gamma-catenin had a distinctive localization to the vertices of these hexagonally shaped differentiating lens fiber cells, a region devoid of actin; while beta-catenin co-localized with actin at lateral cell interfaces. We believe this novel vimentin-linked N-cadherin/gamma-catenin junction provides the tensile strength necessary to establish and maintain structural integrity in tissues that lack desmosomes.


Subject(s)
Cadherins/physiology , Intermediate Filament Proteins/physiology , Lens, Crystalline/cytology , Lens, Crystalline/embryology , gamma Catenin/physiology , Actins/analysis , Actins/physiology , Animals , Cell Differentiation/physiology , Chick Embryo , Epithelial Cells/cytology , Fluorescent Antibody Technique , Vimentin/physiology
15.
Leukemia ; 21(12): 2495-505, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17898786

ABSTRACT

The t(8;21)(q22;q22) occurs frequently in acute myelogenous leukaemia and gives rise to the transcription factor fusion protein, RUNX1-RUNX1T1 (also known as AML1-ETO). To identify the genes dysregulated by the aberrant transcriptional activity of RUNX1-RUNX1T1, we used microarrays to determine the effect of this mutation on gene expression in human progenitor cells and during subsequent development. Gene signatures of these developmental subsets were very dissimilar indicating that effects of RUNX1-RUNX1T1 are highly context dependent. We focused on gene changes associated with the granulocytic lineage and identified a clinically relevant subset of these by comparison with 235 leukaemia patient transcriptional signatures. We confirmed the overexpression of a number of significant genes (Sox4, IL-17BR, CD200 and gamma-catenin). Further, we show that overexpression of CD200 and gamma-catenin is also associated with the inv(16) abnormality which like RUNX1-RUNX1T1 disrupts core binding factor activity. We investigated the functional significance of CD200 and gamma-catenin overexpression in normal human progenitor cells. The effect of IL17 on growth was also assessed. Individually, none of these changes were sufficient to recapitulate the effects of RUNX1-RUNX1T1 on normal development. These data provide the most comprehensive and pertinent assessment of the effect of RUNX1-RUNX1T1 on gene expression and demonstrate the highly context-dependent effects of this fusion gene.


Subject(s)
Core Binding Factor Alpha 2 Subunit/physiology , Gene Expression Regulation, Leukemic/genetics , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/genetics , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/physiology , Transcription, Genetic/genetics , Antigens, CD/biosynthesis , Antigens, CD/genetics , Cell Line, Tumor/metabolism , Cell Lineage , Cells, Cultured/metabolism , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 21/ultrastructure , Chromosomes, Human, Pair 8/genetics , Chromosomes, Human, Pair 8/ultrastructure , Desmoplakins/genetics , Desmoplakins/physiology , Gene Expression Profiling , Hematopoietic Stem Cells/pathology , High Mobility Group Proteins/biosynthesis , High Mobility Group Proteins/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Neoplasm Proteins/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , RUNX1 Translocation Partner 1 Protein , Receptors, Interleukin-17/biosynthesis , Receptors, Interleukin-17/genetics , Recombinant Fusion Proteins/physiology , SOXC Transcription Factors , Trans-Activators/biosynthesis , Trans-Activators/genetics , Translocation, Genetic , gamma Catenin/genetics , gamma Catenin/physiology
16.
Br J Cancer ; 96(12): 1783-7, 2007 Jun 18.
Article in English | MEDLINE | ID: mdl-17519903

ABSTRACT

Much evidence now attests to the importance of desmosomes and their constituents in cancer. Alterations in the expression of desmosomal components could contribute to the progression of the disease by modifying intracellular signal transduction pathways and/or by causing reduced cell adhesion. The Wnt/beta-catenin pathway is a potential target because of the involvement of the cytoplasmic desmosomal protein plakoglobin. Loss of desmosomal adhesion is a prerequisite for the epithelial-mesenchymal transition, implicated in the conversion of early stage tumours to invasive cancers.


Subject(s)
Desmosomes/physiology , Neoplasms/pathology , Cadherins/physiology , Cell Membrane/pathology , Desmosomes/pathology , Humans , Models, Biological , Mutation , Neoplasms/genetics , Neoplasms/physiopathology , Signal Transduction , beta Catenin/physiology , gamma Catenin/physiology
18.
J Invest Dermatol ; 127(4): 792-801, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17110936

ABSTRACT

The armadillo family protein plakoglobin (Pg) is a well-characterized component of anchoring junctions, where it functions to mediate cell-cell adhesion and maintain epithelial tissue integrity. Although its closest homolog beta-catenin acts in the Wnt signaling pathway to dictate cell fate and promote proliferation and survival, the role of Pg in these processes is not well understood. Here, we investigate how Pg affects the survival of mouse keratinocytes by challenging both Pg-null cells and their heterozygote counterparts with apoptotic stimuli. Our results indicate that Pg deletion protects keratinocytes from apoptosis, with null cells exhibiting delayed mitochondrial cytochrome c release and activation of caspase-3. Pg-null keratinocytes also exhibit increased messenger RNA and protein levels of the anti-apoptotic molecule Bcl-X(L) compared to heterozygote controls. Importantly, reintroduction of Pg into the null cells shifts their phenotype towards that of the Pg+/- keratinocytes, providing further evidence that Pg plays a direct role in regulating cell survival. Taken together, our results suggest that in addition to its adhesive role in epithelia, Pg may also function in contrast to the pro-survival tendencies of beta-catenin, to potentiate death in cells damaged by apoptotic stimuli, perhaps limiting the potential for the propagation of mutations and cellular transformation.


Subject(s)
Apoptosis/physiology , Keratinocytes/physiology , gamma Catenin/physiology , Animals , Caspase 3/metabolism , Cell Proliferation , Cells, Cultured , Cytochromes c/metabolism , Enzyme Activation/physiology , Keratinocytes/cytology , Mice , Mice, Knockout , Mitochondria/metabolism , RNA, Messenger/metabolism , Time Factors , bcl-X Protein/genetics , bcl-X Protein/metabolism , gamma Catenin/deficiency
19.
EMBO J ; 25(14): 3298-309, 2006 Jul 26.
Article in English | MEDLINE | ID: mdl-16871158

ABSTRACT

The autoimmune disease pemphigus vulgaris (PV) manifests as loss of keratinocyte cohesion triggered by autoantibody binding to desmoglein (Dsg)3, an intercellular adhesion molecule of mucous membranes, epidermis, and epidermal stem cells. Here we describe a so far unknown signaling cascade activated by PV antibodies. It extends from a transient enhanced turn over of cell surface-exposed, nonkeratin-anchored Dsg3 and associated plakoglobin (PG), through to depletion of nuclear PG, and as one of the consequences, abrogation of PG-mediated c-Myc suppression. In PV patients (6/6), this results in pathogenic c-Myc overexpression in all targeted tissues, including the stem cell compartments. In summary, these results show that PV antibodies act via PG to abolish the c-Myc suppression required for both maintenance of epidermal stem cells in their niche and controlled differentiation along the epidermal lineage. Besides a completely novel insight into PV pathogenesis, these data identify PG as a potent modulator of epithelial homeostasis via its role as a key suppressor of c-Myc.


Subject(s)
Pemphigus/metabolism , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Repressor Proteins/physiology , Skin/metabolism , gamma Catenin/physiology , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Pemphigus/genetics , Proto-Oncogene Proteins c-myc/deficiency , Proto-Oncogene Proteins c-myc/genetics , Repressor Proteins/genetics , beta Catenin/deficiency , beta Catenin/genetics , gamma Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...