Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 313
1.
J Cell Sci ; 136(1)2023 01 01.
Article En | MEDLINE | ID: mdl-36594662

Desmosome diseases are caused by dysfunction of desmosomes, which anchor intermediate filaments (IFs) at sites of cell-cell adhesion. For many decades, the focus of attention has been on the role of actin filament-associated adherens junctions in development and disease, especially cancer. However, interference with the function of desmosomes, their molecular constituents or their attachments to IFs has now emerged as a major contributor to a variety of diseases affecting different tissues and organs including skin, heart and the digestive tract. The first Alpine desmosome disease meeting (ADDM) held in Grainau, Germany, in October 2022 brought together international researchers from the basic sciences with clinical experts from diverse fields to share and discuss their ideas and concepts on desmosome function and dysfunction in the different cell types involved in desmosome diseases. Besides the prototypic desmosomal diseases pemphigus and arrhythmogenic cardiomyopathy, the role of desmosome dysfunction in inflammatory bowel diseases and eosinophilic esophagitis was discussed.


Desmosomes , Disease , Humans , Cell Adhesion , Desmosomes/physiology , Pemphigus
2.
Stem Cell Reports ; 17(2): 337-351, 2022 02 08.
Article En | MEDLINE | ID: mdl-35063130

Loss-of-function mutations in PKP2, which encodes plakophilin-2, cause arrhythmogenic cardiomyopathy (AC). Restoration of deficient molecules can serve as upstream therapy, thereby requiring a human model that recapitulates disease pathology and provides distinct readouts in phenotypic analysis for proof of concept for gene replacement therapy. Here, we generated isogenic induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) with precisely adjusted expression of plakophilin-2 from a patient with AC carrying a heterozygous frameshift PKP2 mutation. After monolayer differentiation, plakophilin-2 deficiency led to reduced contractility, disrupted intercalated disc structures, and impaired desmosome assembly in iPSC-CMs. Allele-specific fluorescent labeling of endogenous DSG2 encoding desmoglein-2 in the generated isogenic lines enabled real-time desmosome-imaging under an adjusted dose of plakophilin-2. Adeno-associated virus-mediated gene replacement of PKP2 recovered contractility and restored desmosome assembly, which was sequentially captured by desmosome-imaging in plakophilin-2-deficient iPSC-CMs. Our isogenic set of iPSC-CMs recapitulates AC pathology and provides a rapid and convenient cellular platform for therapeutic development.


Arrhythmias, Cardiac/pathology , Desmosomes/physiology , Myocardial Contraction/physiology , Plakophilins/metabolism , Arrhythmias, Cardiac/genetics , CRISPR-Cas Systems/genetics , Cell Differentiation , Female , Gene Editing , Genetic Vectors/genetics , Genetic Vectors/metabolism , Heterozygote , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Male , Models, Biological , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Pedigree , Plakophilins/genetics
3.
Annu Rev Pathol ; 17: 47-72, 2022 01 24.
Article En | MEDLINE | ID: mdl-34425055

Desmosomal cadherins are a recent evolutionary innovation that make up the adhesive core of highly specialized intercellular junctions called desmosomes. Desmosomal cadherins, which are grouped into desmogleins and desmocollins, are related to the classical cadherins, but their cytoplasmic domains are tailored for anchoring intermediate filaments instead of actin to sites of cell-cell adhesion. The resulting junctions are critical for resisting mechanical stress in tissues such as the skin and heart. Desmosomal cadherins also act as signaling hubs that promote differentiation and facilitate morphogenesis, creating more complex and effective tissue barriers in vertebrate tissues. Interference with desmosomal cadherin adhesive and supra-adhesive functions leads to a variety of autoimmune, hereditary, toxin-mediated, and malignant diseases. We review our current understanding of how desmosomal cadherins contribute to human health and disease, highlight gaps in our knowledge about their regulation and function, and introduce promising new directions toward combatting desmosome-related diseases.


Desmocollins , Desmosomes , Cadherins/physiology , Cell Adhesion/physiology , Desmosomes/physiology , Humans , Signal Transduction
4.
Curr Biol ; 31(15): 3275-3291.e5, 2021 08 09.
Article En | MEDLINE | ID: mdl-34107301

The epidermis is a stratified epithelium in which structural and functional features are polarized across multiple cell layers. This type of polarity is essential for establishing the epidermal barrier, but how it is created and sustained is poorly understood. Previous work identified a role for the classic cadherin/filamentous-actin network in establishment of epidermal polarity. However, little is known about potential roles of the most prominent epidermal intercellular junction, the desmosome, in establishing epidermal polarity, in spite of the fact that desmosome constituents are patterned across the apical to basal cell layers. Here, we show that desmosomes and their associated intermediate filaments (IFs) are key regulators of mechanical polarization in epidermis, whereby basal and suprabasal cells experience different forces that drive layer-specific functions. Uncoupling desmosomes and IF or specific targeting of apical desmosomes through depletion of the superficial desmosomal cadherin, desmoglein 1, impedes basal stratification in an in vitro competition assay and suprabasal tight junction barrier functions in 3D reconstructed epidermis. Surprisingly, disengaging desmosomes from IF also accelerated the expression of differentiation markers, through precocious activation of the mechanosensitive transcriptional regulator serum response factor (SRF) and downstream activation of epidermal growth factor receptor family member ErbB2 by Src family kinase (SFK)-mediated phosphorylation. This Dsg1-SFK-ErbB2 axis also helps maintain tight junctions and barrier function later in differentiation. Together, these data demonstrate that the desmosome-IF network is a critical contributor to the cytoskeletal-adhesive machinery that supports the polarized function of the epidermis.


Desmosomes , Epidermis , Cadherins , Desmoplakins , Desmosomes/physiology , Epidermal Cells , Epidermis/physiology
5.
J Invest Dermatol ; 141(5): 1219-1229.e11, 2021 05.
Article En | MEDLINE | ID: mdl-33098828

Intercellular adhesion is essential for tissue integrity and homeostasis. Desmosomes are abundant in the epidermis and the myocardium-tissues, which are under constantly changing mechanical stresses. Yet, it is largely unclear whether desmosomal adhesion can be rapidly adapted to changing demands, and the mechanisms underlying desmosome turnover are only partially understood. In this study we show that the loss of the actin-binding protein α-adducin resulted in reduced desmosome numbers and prevented the ability of cultured keratinocytes or murine epidermis to withstand mechanical stress. This effect was not primarily caused by decreased levels or impaired adhesive properties of desmosomal molecules but rather by altered desmosome turnover. Mechanistically, reduced cortical actin density in α-adducin knockout keratinocytes resulted in increased mobility of the desmosomal adhesion molecule desmoglein 3 and impaired interactions with E-cadherin, a crucial step in desmosome formation. Accordingly, the loss of α-adducin prevented increased membrane localization of desmoglein 3 in response to cyclic stretch or shear stress. Our data demonstrate the plasticity of desmosomal molecules in response to mechanical stimuli and unravel a mechanism of how the actin cytoskeleton indirectly shapes intercellular adhesion by restricting the membrane mobility of desmosomal molecules.


Calmodulin-Binding Proteins/physiology , Desmosomes/physiology , Microfilament Proteins/physiology , Animals , Cadherins/chemistry , Calcium/metabolism , Cell Adhesion , Cell Plasticity , Cells, Cultured , Desmoglein 3/metabolism , Desmosomes/chemistry , Humans , Mice
7.
Proc Natl Acad Sci U S A ; 117(44): 27132-27140, 2020 11 03.
Article En | MEDLINE | ID: mdl-33067392

Desmosomes are cell-cell junctions that link tissue cells experiencing intense mechanical stress. Although the structure of the desmosomal cadherins is known, the desmosome architecture-which is essential for mediating numerous functions-remains elusive. Here, we recorded cryo-electron tomograms (cryo-ET) in which individual cadherins can be discerned; they appear variable in shape, spacing, and tilt with respect to the membrane. The resulting sub-tomogram average reaches a resolution of ∼26 Å, limited by the inherent flexibility of desmosomes. To address this challenge typical of dynamic biological assemblies, we combine sub-tomogram averaging with atomistic molecular dynamics (MD) simulations. We generate models of possible cadherin arrangements and perform an in silico screening according to biophysical and structural properties extracted from MD simulation trajectories. We find a truss-like arrangement of cadherins that resembles the characteristic footprint seen in the electron micrograph. The resulting model of the desmosomal architecture explains their unique biophysical properties and strength.


Desmosomes/chemistry , Electron Microscope Tomography/methods , Cadherins/chemistry , Cadherins/metabolism , Desmosomes/metabolism , Desmosomes/physiology , Humans , Intercellular Junctions , Molecular Dynamics Simulation
8.
Mol Biol Cell ; 31(11): 1140-1153, 2020 05 15.
Article En | MEDLINE | ID: mdl-32238101

Desmosomes are cell-cell adhesions necessary for the maintenance of tissue integrity in the skin and heart. While the core components of desmosomes have been identified, peripheral components that modulate canonical or noncanonical desmosome functions still remain largely unexplored. Here we used targeted proximity labeling approaches to further elaborate the desmosome proteome in epidermal keratinocytes. Quantitative mass spectrometry analysis identified all core desmosomal proteins while uncovering a diverse array of new constituents with broad molecular functions. By individually targeting the inner and outer dense plaques, we defined proteins enriched within these subcompartments. We validated a number of these novel desmosome-associated proteins and find that many are membrane proximal proteins that show a dependence on functional desmosomes for their cortical localization. We further explored the mechanism of localization and function of two novel desmosome-associated adaptor proteins enriched in the desmosome proteome, Crk and Crk-like (CrkL). These proteins interacted with Dsg1 and rely on Dsg1 and desmoplakin for robust cortical localization. Epidermal deletion of both Crk and CrkL resulted in perinatal lethality with defects in desmosome morphology and keratin organization, thus demonstrating the utility of this dataset in identifying novel proteins required for desmosome-dependent epidermal integrity.


Desmosomes/metabolism , Epidermis/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Adhesion , Cytoskeleton/metabolism , Desmoplakins/metabolism , Desmosomes/physiology , Epidermal Cells/metabolism , Epidermal Cells/physiology , Epidermis/physiology , Humans , Keratinocytes/metabolism , Keratins/metabolism , Male , Mass Spectrometry/methods , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Proteomics/methods , Proto-Oncogene Proteins c-crk/metabolism , Skin/metabolism
9.
Mol Biol Cell ; 31(8): 741-752, 2020 04 01.
Article En | MEDLINE | ID: mdl-32049581

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.


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
10.
J Invest Dermatol ; 139(11): 2292-2301.e9, 2019 11.
Article En | MEDLINE | ID: mdl-31129056

Cornifelin (CNFN) has been identified as a protein component of epidermal corneocytes. Here, we investigated the tissue distribution of CNFN and potential consequences of CNFN deficiency on epithelial function in in vitro models of human skin and oral mucosa. Our detailed bioinformatics and immunostaining analysis revealed that CNFN is not only expressed in human epidermis but also in noncornifying oral mucosa. In normal epidermis, CNFN was confined to the upper granular layer and the stratum corneum. By contrast, in both partly cornifying and noncornifying oral mucosa, CNFN was expressed in a cell membrane-associated pattern over several suprabasal layers. Small interfering RNA-mediated knockdown of CNFN in epidermal keratinocytes (KCs) was associated with only subtle alterations of the overall epidermal architecture in skin models in vitro but led to altered morphology of corneodesmosomes, as detected by electron microscopy. Using dispase treatment followed by mechanical stress, epithelial sheets of CNFN-deficient epidermal KCs were easily disrupted, whereas their CNFN-competent counterparts remained intact. In contrast to the epidermal KCs, CNFN knockdown in oral KCs had a more severe effect and caused pronounced acantholysis in organotypic models of oral mucosa. Together, these findings indicate that CNFN is a structural component of the cell adhesion system of differentiated KCs in both epidermis and oral mucosa.


Acantholysis/genetics , Desmosomes/physiology , Epidermis/pathology , Keratinocytes/physiology , Membrane Proteins/metabolism , Mouth Mucosa/pathology , Cell Adhesion , Cell Differentiation , Cells, Cultured , Desmogleins/metabolism , Epidermis/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Mouth Mucosa/metabolism , Organ Culture Techniques , RNA, Small Interfering/genetics
11.
F1000Res ; 82019.
Article En | MEDLINE | ID: mdl-31942240

The development of adhesive connections between cells was critical for the evolution of multicellularity and for organizing cells into complex organs with discrete compartments. Four types of intercellular junction are present in vertebrates: desmosomes, adherens junctions, tight junctions, and gap junctions. All are essential for the development of the embryonic layers and organs as well as adult tissue homeostasis. While each junction type is defined as a distinct entity, it is now clear that they cooperate physically and functionally to create a robust and functionally diverse system. During evolution, desmosomes first appeared in vertebrates as highly specialized regions at the plasma membrane that couple the intermediate filament cytoskeleton at points of strong cell-cell adhesion. Here, we review how desmosomes conferred new mechanical and signaling properties to vertebrate cells and tissues through their interactions with the existing junctional and cytoskeletal network.


Cytoskeleton/physiology , Desmosomes/physiology , Intercellular Junctions/physiology , Animals , Signal Transduction
12.
Ann Anat ; 226: 96-100, 2019 Nov.
Article En | MEDLINE | ID: mdl-30529571

Cell-cell contacts are crucial for intercellular cohesion and formation of endothelial and epithelial barriers. Desmosomes are the adhesive contacts providing mechanical strength to epithelial intercellular adhesion and therefore are most abundant in tissues subjected to high mechanical stress such as the epidermis and heart muscle. Desmogleins (Dsg) besides intercellular adhesion serve as signalling hubs regulating cell behaviour. In desmosomal diseases such as the autoimmune blistering skin disease pemphigus or arrhythmic cardiomyopathy (AC), which is caused by mutations of desmosomal components of cardiomyocyte intercalated discs, the adhesive and signalling functions of desmosomes are impaired. Therefore, our goal is to elucidate the mechanisms regulating adhesion of desmosomes in order to develop new strategies to treat desmosomal diseases. For pemphigus, we have provided evidence that intracellular signalling is required for loss of keratinocyte cohesion and have characterized a first disease-relevant adhesion receptor consisting of Dsg3 and p38MAPK. We propose that signalling patterns correlate with autoantibody profiles and thereby define the clinical phenotypes of pemphigus. Besides direct modulation of signalling pathways we have demonstrated that peptide-mediated crosslinking of Dsg molecules can abolish skin blistering in vivo. A similar approach may be effective to stabilize adhesion in cardiomyocytes of AC hearts. Since we observed that the adrenergic ß1-receptor is localized at intercalated discs we evaluate signalling pathways regulating cardiomyocyte cohesion. With adrenergic signalling we have reported a first mechanism to stabilize desmosomal adhesion in intercalated discs and proposed a new function of the sympathicus in the heart we refer to as positive adhesiotropy.


Desmogleins/physiology , Heart/physiology , Myocardium/cytology , Signal Transduction/physiology , Skin Physiological Phenomena , Skin/cytology , Desmosomes/physiology , Epidermal Cells , Humans
13.
Ann Biomed Eng ; 47(3): 852-865, 2019 Mar.
Article En | MEDLINE | ID: mdl-30569242

Cell junctions play an important role in coordinating intercellular communication and intracellular ultrastructures, with desmosomes representing the mechanical component of such intercellular connections. Mutations to desmosomal component proteins compromise both inter- and intracellular signalling and correlate with severe diseases like arrhythmogenic cardiomyopathy (AC), with pathological phenotypes in tissues subjected to intense mechanical stimuli (skin and heart). Here, we explore the consequences of dysfunctional desmosomes in one line of induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) derived from an AC patient with a homozygous pathogenic mutation in desmosomal component protein plakophilin-2 (PKP2). We specifically aim at investigating the response to mechanical stress in an AC-pathological setting. To this aim, we aligned hiPS-CMs on stretchable patterned substrates to mimic the cardiac functional syncytium and compared transcriptomic profiles of PKP2-mutated hiPS-CMs and healthy controls. AC-CMs display altered transcription towards a pro-fibrotic gene expression program, and concurrent dysregulation of gene sets closely associated with cell-to-cell connections. By integrating the culture substrate with a macroscopic stretching setup able to accurately apply cyclic uniaxial elongation, we show how response to mechanical loads in AC-CMs deviates from the canonical mechanical-stress response observed in healthy-CMs.


Arrhythmias, Cardiac/physiopathology , Cardiomyopathies/physiopathology , Myocytes, Cardiac/physiology , Body Patterning , Desmosomes/physiology , Finite Element Analysis , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/cytology , Physical Stimulation , Plakophilins/genetics , Stress, Mechanical
14.
Mucosal Immunol ; 11(6): 1630-1639, 2018 11.
Article En | MEDLINE | ID: mdl-30115995

Desmosomes are the least understood intercellular junctions in the intestinal epithelia and provide cell-cell adhesion via the cadherins desmoglein (Dsg)2 and desmocollin (Dsc)2. We studied these cadherins in Crohn's disease (CD) patients and in newly generated conditional villin-Cre DSG2 and DSC2 knockout mice (DSG2ΔIEC; DSC2ΔIEC). CD patients exhibited altered desmosomes and reduced Dsg2/Dsc2 levels. The intestines of both transgenic animal lines were histopathologically inconspicuous. However, DSG2ΔIEC, but not DSC2ΔIEC mice displayed an increased intestinal permeability, a wider desmosomal space as well as alterations in desmosomal and tight junction components. After dextran sodium sulfate (DSS) treatment and Citrobacter rodentium exposure, DSG2ΔIEC mice developed a more-pronounced colitis, an enhanced intestinal epithelial barrier disruption, leading to a stronger inflammation and activation of epithelial pSTAT3 signaling. No susceptibility to DSS-induced intestinal injury was noted in DSC2ΔIEC animals. Dsg2 interacted with the cytoprotective chaperone Hsp70. Accordingly, DSG2ΔIEC mice had lower Hsp70 levels in the plasma membrane compartment, whereas DSC2ΔIEC mice displayed a compensatory recruitment of galectin 3, a junction-tightening protein. Our results demonstrate that Dsg2, but not Dsc2 is required for the integrity of the intestinal epithelial barrier in vivo.


Crohn Disease/immunology , Desmoglein 2/metabolism , Desmosomes/physiology , Intestinal Mucosa/physiology , Membrane Glycoproteins/metabolism , Adult , Aged , Animals , Cell Adhesion , Desmocollins , Desmoglein 2/genetics , Galectin 3/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Young Adult
15.
J Cell Biol ; 217(9): 3219-3235, 2018 09 03.
Article En | MEDLINE | ID: mdl-29959233

Desmoplakin (DP) is an obligate component of desmosomes, intercellular adhesive junctions that maintain the integrity of the epidermis and myocardium. Mutations in DP can cause cardiac and cutaneous disease, including arrhythmogenic cardiomyopathy (ACM), an inherited disorder that frequently results in deadly arrhythmias. Conduction defects in ACM are linked to the remodeling and functional interference with Cx43-based gap junctions that electrically and chemically couple cells. How DP loss impairs gap junctions is poorly understood. We show that DP prevents lysosomal-mediated degradation of Cx43. DP loss triggered robust activation of ERK1/2-MAPK and increased phosphorylation of S279/282 of Cx43, which signals clathrin-mediated internalization and subsequent lysosomal degradation of Cx43. RNA sequencing revealed Ras-GTPases as candidates for the aberrant activation of ERK1/2 upon loss of DP. Using a novel Ras inhibitor, Ras/Rap1-specific peptidase (RRSP), or K-Ras knockdown, we demonstrate restoration of Cx43 in DP-deficient cardiomyocytes. Collectively, our results reveal a novel mechanism for the regulation of the Cx43 life cycle by DP in cardiocutaneous models.


Connexin 43/metabolism , Desmoplakins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gap Junctions/physiology , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Cardiomyopathies/pathology , Cell Communication/physiology , Cells, Cultured , Clathrin/metabolism , Desmoplakins/genetics , Desmosomes/physiology , Enzyme Activation/genetics , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Rats , Rats, Sprague-Dawley
16.
J Dermatol Sci ; 89(3): 241-247, 2018 Mar.
Article En | MEDLINE | ID: mdl-29198699

BACKGROUND: Mechanical stress is an ubiquitous challenge of human cells with fundamental impact on cell physiology. Previous studies have shown that stretching promotes signalling cascades involved in proliferation and tissue enlargement. OBJECTIVE: The present study is dedicated to learn more about cellular structures contributing to perception and signal transmission of cell stretch. In particular, we hypothesized that desmosmal contacts and the adjacent keratin filament build an intercellular matrix providing information about the mechanical load. METHODS: Epidermal cells with different keratin equipment were seeded on flexible silicon dishes and stretched. As read out parameter the activation of PKB/Akt and p44/42 was monitored by Western blotting. Likewise desomosomal contacts were manipulated by depletion or addition of calcium. Moreover, desmoglein 3 and desmocollin 3 were blocked by either specific antibodies or siRNA. RESULTS: It was found that the omission of calcium from the medium, a necessary cofactor for desmosomal cadherins, inhibited stretch mediated activation of PKB/Akt and p44/42. The relevance of desmosomes in this context was further substantiated by experiments using a desmoglein 3 blocking antibody (AK23) and siRNA against desmocollin 3. Moreover, disruption of the keratin filament by sodium orthovanadate also abrogates PKB/Akt and p44/42 activation in response to stretch. Likewise, KEB-7 keratinocytes harbouring a mutation in the keratin 14 gene and genetically modified keratinocytes devoid of any keratin show an altered signalling after stretch indicating the relevance of the keratin filament in this context. CONCLUSION: Besides their important role in cell architecture our results identify desmosomes and keratins as mechanosensing structures.


Desmosomes/physiology , Keratins/physiology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Proto-Oncogene Proteins c-akt/physiology , Calcium/physiology , Cells, Cultured , Desmoglein 3/physiology , Enzyme Activation , Humans , Stress, Mechanical
17.
Article En | MEDLINE | ID: mdl-28600395

Cell-cell junctions link cells to each other in tissues, and regulate tissue homeostasis in critical cell processes that include tissue barrier function, cell proliferation, and migration. Defects in cell-cell junctions give rise to a wide range of tissue abnormalities that disrupt homeostasis and are common in genetic abnormalities and cancers. Here, we discuss the organization and function of cell-cell junctions primarily involved in adhesion (tight junction, adherens junction, and desmosomes) in two different epithelial tissues: a simple epithelium (intestine) and a stratified epithelium (epidermis). Studies in these tissues reveal similarities and differences in the organization and functions of different cell-cell junctions that meet the requirements for the specialized functions of each tissue. We discuss cell-cell junction responses to genetic and environmental perturbations that provide further insights into their roles in maintaining tissue homeostasis.


Epithelial Cells/cytology , Intercellular Junctions/physiology , Adherens Junctions/physiology , Animals , Cell Movement , Cell Proliferation , Desmosomes/physiology , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Epithelium/metabolism , Homeostasis , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiology , Signal Transduction , Tight Junctions/physiology , Wound Healing
18.
Biochem Biophys Res Commun ; 495(1): 768-774, 2018 01 01.
Article En | MEDLINE | ID: mdl-29146182

Previous reports show that the desmosomal plaque protein plakophilin3 (PKP3) is essential for desmosome formation. Here, we report that PKP3 over-expression decreases calcium dependency for de novo desmosome formation and makes existing cell-cell adhesion junctions more resilient in low calcium medium due to an increase in desmocollin2 expression. PKP3 overexpression increases the stability of other desmosomal proteins independently of the increase in DSC2 levels and regulates desmosome formation and stability by a multimodal mechanism affecting transcription, protein stability and cell border localization of desmosomal proteins.


Cell Adhesion/physiology , Desmocollins/metabolism , Desmosomes/physiology , Desmosomes/ultrastructure , Plakophilins/metabolism , Cell Line , Humans , Particle Size
19.
Article En | MEDLINE | ID: mdl-28893859

Cadherin-based adherens junctions (AJs) and desmosomes are crucial to couple intercellular adhesion to the actin or intermediate filament cytoskeletons, respectively. As such, these intercellular junctions are essential to provide not only integrity to epithelia and other tissues but also the mechanical machinery necessary to execute complex morphogenetic and homeostatic intercellular rearrangements. Moreover, these spatially defined junctions serve as signaling hubs that integrate mechanical and chemical pathways to coordinate tissue architecture with behavior. This review takes an evolutionary perspective on how the emergence of these two essential intercellular junctions at key points during the evolution of multicellular animals afforded metazoans with new opportunities to integrate adhesion, cytoskeletal dynamics, and signaling. We discuss known literature on cross-talk between the two junctions and, using the skin epidermis as an example, provide a model for how these two junctions function in concert to orchestrate tissue organization and function.


Adherens Junctions/physiology , Biological Evolution , Desmosomes/physiology , Signal Transduction/physiology , Adherens Junctions/genetics , Animals , Cell Polarity , Desmosomes/genetics , Epithelial Cells/physiology
20.
J Clin Invest ; 127(11): 4001-4017, 2017 Nov 01.
Article En | MEDLINE | ID: mdl-28945203

Despite its central position in oncogenic intracellular signaling networks, the role of mTORC1 in epithelial development has not been studied extensively in vivo. Here, we have used the epidermis as a model system to elucidate the cellular effects and signaling feedback sequelae of mTORC1 loss of function in epithelial tissue. In mice with conditional epidermal loss of the mTORC1 components Rheb or Rptor, mTORC1 loss of function unexpectedly resulted in a profound skin barrier defect with epidermal abrasions, blistering, and early postnatal lethality, due to a thinned epidermis with decreased desmosomal protein expression and incomplete biochemical differentiation. In mice with mTORC1 loss of function, we found that Rho kinase (ROCK) signaling was constitutively activated, resulting in increased cytoskeletal tension and impaired cell-cell adhesion. Inhibition or silencing of ROCK1 was sufficient to rescue keratinocyte adhesion and biochemical differentiation in these mice. mTORC1 loss of function also resulted in marked feedback upregulation of upstream TGF-ß signaling, triggering ROCK activity and its downstream effects on desmosomal gene expression. These findings elucidate a role for mTORC1 in the regulation of epithelial barrier formation, cytoskeletal tension, and cell adhesion, underscoring the complexity of signaling feedback following mTORC1 inhibition.


Keratinocytes/physiology , Mechanistic Target of Rapamycin Complex 1/genetics , Signal Transduction , Animals , Cell Adhesion , Cell Differentiation , Cells, Cultured , Desmosomes/physiology , Enzyme Activation , Epidermal Cells , Female , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , rho-Associated Kinases/metabolism
...