Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 210
Filter
1.
J Invest Dermatol ; 142(2): 323-332.e8, 2022 02.
Article in English | MEDLINE | ID: mdl-34352264

ABSTRACT

Dominant and recessive mutations in the desmosomal cadherin, desmoglein (DSG) 1, cause the skin diseases palmoplantar keratoderma (PPK) and severe dermatitis, multiple allergies, and metabolic wasting (SAM) syndrome, respectively. In this study, we compare two dominant missense mutations in the DSG1 transmembrane domain (TMD), G557R and G562R, causing PPK (DSG1PPK-TMD) and SAM syndrome (DSG1SAM-TMD), respectively, to determine the differing pathomechanisms of these mutants. Expressing the DSG1TMD mutants in a DSG-null background, we use cellular and biochemical assays to reveal the differences in the mechanistic behavior of each mutant. Super-resolution microscopy and functional assays showed a failure by both mutants to assemble desmosomes due to reduced membrane trafficking and lipid raft targeting. DSG1SAM-TMD maintained normal expression levels and turnover relative to wildtype DSG1, but DSG1PPK-TMD lacked stability, leading to increased turnover through lysosomal and proteasomal pathways and reduced expression levels. These results differentiate the underlying pathomechanisms of these disorders, suggesting that DSG1SAM-TMD acts dominant negatively, whereas DSG1PPK-TMD is a loss-of-function mutation causing the milder PPK disease phenotype. These mutants portray the importance of the DSG TMD in desmosome function and suggest that a greater understanding of the desmosomal cadherin TMDs will further our understanding of the role that desmosomes play in epidermal pathophysiology.


Subject(s)
Desmoglein 1/genetics , Desmosomes/pathology , Epidermis/pathology , Keratoderma, Palmoplantar/genetics , Cell Adhesion/genetics , Cell Line, Tumor , Desmoglein 1/metabolism , Desmosomal Cadherins/metabolism , Desmosomes/metabolism , Epidermis/metabolism , Humans , Keratoderma, Palmoplantar/pathology , Loss of Function Mutation , Membrane Microdomains/metabolism , Mutation, Missense , Protein Domains/genetics , Protein Stability
2.
Nat Commun ; 12(1): 6795, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34815391

ABSTRACT

Eosinophilic esophagitis (EoE) is a chronic allergic inflammatory disease with a complex underlying genetic etiology. Herein, we conduct whole-exome sequencing of a multigeneration EoE pedigree (discovery set) and 61 additional multiplex families with EoE (replication set). A series of rare, heterozygous, missense variants are identified in the genes encoding the desmosome-associated proteins DSP and PPL in 21% of the multiplex families. Esophageal biopsies from patients with these variants retain dilated intercellular spaces and decrease DSP and PPL expression even during disease remission. These variants affect barrier integrity, cell motility and RhoGTPase activity in esophageal epithelial cells and have increased susceptibility to calpain-14-mediated degradation. An acquired loss of esophageal DSP and PPL is present in non-familial EoE. Taken together, herein, we uncover a pathogenic role for desmosomal dysfunction in EoE, providing a deeper mechanistic understanding of tissue-specific allergic responses.


Subject(s)
Desmoplakins/genetics , Eosinophilic Esophagitis/genetics , Esophageal Mucosa/pathology , Plakins/genetics , Adolescent , Biopsy , Calpain/metabolism , Case-Control Studies , Child , DNA Mutational Analysis , Desmoplakins/metabolism , Desmosomes/pathology , Eosinophilic Esophagitis/pathology , Esophageal Mucosa/cytology , Female , HEK293 Cells , HaCaT Cells , Heterozygote , Humans , Male , Mutation, Missense , Plakins/metabolism , Proteolysis , RNA-Seq , Single-Cell Analysis , Exome Sequencing
3.
Basic Res Cardiol ; 116(1): 39, 2021 06 04.
Article in English | MEDLINE | ID: mdl-34089132

ABSTRACT

Arrhythmogenic cardiomyopathy (AC) is an inherited disease characterized by progressive breakdown of heart muscle, myocardial tissue death, and fibrofatty replacement. In most cases of AC, the primary lesion occurs in one of the genes encoding desmosomal proteins, disruption of which increases membrane fragility at the intercalated disc. Disrupted, exposed desmosomal proteins also serve as epitopes that can trigger an autoimmune reaction. Damage to cell membranes and autoimmunity provoke myocardial inflammation, a key feature in early stages of the disease. In several preclinical models, targeting inflammation has been shown to blunt disease progression, but translation to the clinic has been sparse. Here we review current understanding of inflammatory pathways and how they interact with injured tissue and the immune system in AC. We further discuss the potential role of immunomodulatory therapies in AC.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia/metabolism , Desmosomes/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Myocardium/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Arrhythmogenic Right Ventricular Dysplasia/immunology , Arrhythmogenic Right Ventricular Dysplasia/pathology , Arrhythmogenic Right Ventricular Dysplasia/therapy , Cell- and Tissue-Based Therapy , Desmosomes/drug effects , Desmosomes/immunology , Desmosomes/pathology , Genetic Therapy , Humans , Immunomodulating Agents/pharmacology , Immunotherapy , Inflammation/immunology , Inflammation/pathology , Inflammation/therapy , Inflammation Mediators/antagonists & inhibitors , Myocardium/immunology , Myocardium/pathology , Signal Transduction
4.
J Clin Invest ; 131(11)2021 06 01.
Article in English | MEDLINE | ID: mdl-33857019

ABSTRACT

Dysregulated protein degradative pathways are increasingly recognized as mediators of human disease. This mechanism may have particular relevance to desmosomal proteins that play critical structural roles in both tissue architecture and cell-cell communication, as destabilization/breakdown of the desmosomal proteome is a hallmark of genetic-based desmosomal-targeted diseases, such as the cardiac disease arrhythmogenic right ventricular dysplasia/cardiomyopathy (ARVD/C). However, no information exists on whether there are resident proteins that regulate desmosomal proteome homeostasis. Here, we uncovered a cardiac constitutive photomorphogenesis 9 (COP9) desmosomal resident protein complex, composed of subunit 6 of the COP9 signalosome (CSN6), that enzymatically restricted neddylation and targeted desmosomal proteome degradation. CSN6 binding, localization, levels, and function were affected in hearts of classic mouse and human models of ARVD/C affected by desmosomal loss and mutations, respectively. Loss of desmosomal proteome degradation control due to junctional reduction/loss of CSN6 and human desmosomal mutations destabilizing junctional CSN6 were also sufficient to trigger ARVD/C in mice. We identified a desmosomal resident regulatory complex that restricted desmosomal proteome degradation and disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Arrhythmogenic Right Ventricular Dysplasia/metabolism , COP9 Signalosome Complex/metabolism , Desmosomes/metabolism , Proteolysis , Proteome/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Arrhythmogenic Right Ventricular Dysplasia/genetics , COP9 Signalosome Complex/genetics , Desmosomes/genetics , Desmosomes/pathology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Knockout , Proteome/genetics
6.
Cancer Res ; 81(6): 1513-1527, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33461973

ABSTRACT

Ras proteins play a causal role in human cancer by activating multiple pathways that promote cancer growth and invasion. However, little is known about how Ras induces the first diagnostic features of invasion in solid tumors, including loss of epithelial integrity and breaching of the basement membrane (BM). In this study, we found that oncogenic Ras strongly promotes the activation of hepsin, a member of the hepsin/TMPRSS type II transmembrane serine protease family. Mechanistically, the Ras-dependent hepsin activation was mediated via Raf-MEK-ERK signaling, which controlled hepsin protein stability through the heat shock transcription factor-1 stress pathway. In Ras-transformed three-dimensional mammary epithelial culture, ablation of hepsin restored desmosomal cell-cell junctions, hemidesmosomes, and BM integrity and epithelial cohesion. In tumor xenografts harboring mutant KRas, silencing of hepsin increased local invasion concomitantly with accumulation of collagen IV. These findings suggest that hepsin is a critical protease for Ras-dependent tumorigenesis, executing cell-cell and cell-matrix pathologies important for early tumor dissemination. SIGNIFICANCE: These findings identify the cell-surface serine protease hepsin as a potential therapeutic target for its role in oncogenic Ras-mediated deregulation of epithelial cell-cell and cell-matrix interactions and cohesion of epithelial structure.


Subject(s)
Breast Neoplasms/pathology , Epithelial Cells/pathology , Heat Shock Transcription Factors/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Serine Endopeptidases/metabolism , Animals , Basement Membrane/cytology , Basement Membrane/pathology , Breast/pathology , Breast Neoplasms/genetics , Carcinogenesis/pathology , Cell Communication , Cell Line, Tumor , Collagen Type IV/metabolism , Desmosomes/pathology , Epithelial Cells/cytology , Female , Gene Knockdown Techniques , Heat Shock Transcription Factors/genetics , Humans , MAP Kinase Signaling System/genetics , Mammary Glands, Animal/cytology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Mutation , Neoplasm Invasiveness/pathology , Primary Cell Culture , Protein Stability , Proto-Oncogene Proteins p21(ras)/genetics , Serine Endopeptidases/genetics , Up-Regulation , Xenograft Model Antitumor Assays
7.
Int J Mol Sci ; 23(1)2021 Dec 21.
Article in English | MEDLINE | ID: mdl-35008484

ABSTRACT

In arrhythmogenic cardiomyopathy (ACM) pathogenic variants are found in genes encoding desmosomal proteins and in non-desmosomal genes, such as phospholamban (PLN, p.Arg14del variant). Previous research showed that plakoglobin protein levels and localization in the cardiac tissue of ACM patients, and PLN p.Arg14del patients diagnosed with an ACM phenotype, are disturbed. Moreover, the effects of pathogenic variants in desmosomal genes are reflected in non-cardiac tissues like buccal mucosa cells (BMC) which could serve as a promising new and non-invasive tool to support diagnosis. We collected the BMC of 33 ACM patients, 17 PLN p.Arg14del patients and 34 controls, labelled the BMC with anti-plakoglobin antibodies at different concentrations, and scored their membrane labelling. We found that plakoglobin protein levels were significantly reduced in BMC obtained from diagnosed ACM patients and preclinical variant carriers when compared to controls. This effect was independent from age and sex. Moderate to strong correlations were found with the revised 2010 Task Force Criteria score which is commonly used for ACM diagnosis (rs = -0.67, n = 64, p < 0.0001 and rs = -0.71, n = 64, p < 0.0001). In contrast, plakoglobin scores in PLN p.Arg14del patients were comparable to controls (p > 0.209), which suggests differences in underlying etiology. However, for the individual diagnosis of the 'classical' ACM patient, this method might not be discriminative enough to distinguish true patients from variant carriers and controls, because of the high interindividual variability.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia/diagnosis , Arrhythmogenic Right Ventricular Dysplasia/pathology , Mouth Mucosa/pathology , Adult , Arrhythmogenic Right Ventricular Dysplasia/metabolism , Calcium-Binding Proteins/metabolism , Desmosomes/metabolism , Desmosomes/pathology , Disease Progression , Female , Humans , Male , Middle Aged , Mouth Mucosa/metabolism , gamma Catenin/metabolism
8.
Signal Transduct Target Ther ; 5(1): 249, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33122631

ABSTRACT

Novel effective treatment is direly needed for patients with pancreatic ductal adenocarcinoma (PDAC). Therapeutics that target the driver mutations, especially the KRAS oncoprotein and its effector cascades, have been ineffective. It is increasing clear that the extensive fibro-inflammatory stroma (or desmoplasia) of PDAC plays an active role in the progression and therapeutic resistance of PDAC. The desmoplastic stroma is composed of dense extracellular matrix (ECM) deposited mainly by the cancer-associated-fibroblasts (CAFs) and infiltrated with various types of immune cells. The dense ECM functions as a physical barrier that limits tumor vasculatures and distribution of therapeutics to PDAC cells. In addition, mounting evidence have demonstrated that both CAFs and ECM promote PDAC cells aggressiveness through multiple mechanisms, particularly engagement of the epithelial-mesenchymal transition (EMT) program. Acquisition of a mesenchymal-like phenotype renders PDAC cells more invasive and resistant to therapy-induced apoptosis. Here, we critically review seminal and recent articles on the signaling mechanisms by which each stromal element promotes EMT in PDAC. We discussed the experimental models that are currently employed and best suited to study EMT in PDAC, which are instrumental in increasing the chance of successful clinical translation.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Desmosomes/metabolism , Epithelial-Mesenchymal Transition , Pancreatic Neoplasms/metabolism , Signal Transduction , Animals , Carcinoma, Pancreatic Ductal/blood supply , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/therapy , Desmosomes/pathology , Humans , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy
9.
Oncol Rep ; 44(3): 1105-1115, 2020 09.
Article in English | MEDLINE | ID: mdl-32705241

ABSTRACT

Paxillin (PXN) is a cytoplasmic protein that plays an important role in regulating focal adhesion, cytoskeletal rearrangements and cell motility. The present study aimed to investigate the role of PXN in the metastasis of human colorectal cancer (CRC) and its possible mechanisms. Immunohistochemical staining of tissues from 102 surgical CRC patients revealed that high PXN expression was positively correlated with tumour­node­metastasis (TNM) stage, lymph node metastasis, distant metastasis, and recurrence at distant sites after radical surgery. In 24 cases of stage IV CRC, PXN expression in liver metastasis was higher than that in the matched primary tumour. The knockdown of PXN inhibited the proliferation, migration and invasion potential of SW480 cells in vitro and in vivo. Transmission electron microscopy revealed the effect of PXN on ultrastructural characteristics, observed mainly in microvilli and desmosomes. The downregulation of PXN decreased the activation of extracellular regulated protein kinase (ERK) and suppressed the epithelial­mesenchymal transition (EMT) process. Following the downregulation of PXN, the addition of an ERK activator or inhibitor restored or further suppressed EMT, respectively, accompanied by corresponding changes in cell migration and invasion. Collectively, the present results confirmed the important role of PXN in CRC metastasis and revealed that PXN regulated EMT progression via the ERK signalling pathway. PXN may represent a future therapeutic strategy to prevent the EMT­associated progression and invasion of CRC.


Subject(s)
Colorectal Neoplasms/pathology , Liver Neoplasms/secondary , Lymphatic Metastasis/pathology , Neoplasm Recurrence, Local/epidemiology , Paxillin/metabolism , Aged , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation/drug effects , Colon/pathology , Colon/surgery , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/surgery , Desmosomes/pathology , Desmosomes/ultrastructure , Disease Progression , Epithelial-Mesenchymal Transition/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Follow-Up Studies , Humans , Intestinal Mucosa/pathology , Intestinal Mucosa/surgery , Liver/pathology , MAP Kinase Signaling System/drug effects , Male , Mice , Microscopy, Electron, Transmission , Microvilli/pathology , Microvilli/ultrastructure , Middle Aged , Neoplasm Invasiveness/pathology , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/prevention & control , Paxillin/genetics , Retrospective Studies , Xenograft Model Antitumor Assays
10.
J Invest Dermatol ; 140(2): 319-326.e4, 2020 02.
Article in English | MEDLINE | ID: mdl-31356814

ABSTRACT

The skin permeability barrier is indispensable for maintaining water inside the body and preventing the invasion of pathogens and allergens; abnormalities lead to skin disorders such as atopic dermatitis and ichthyosis. Acylceramide is an essential lipid for skin barrier formation, and CYP4F22 is a fatty acid ω-hydroxylase involved in its synthesis. Mutations in CYP4F22 cause autosomal recessive congenital ichthyosis, although the symptoms vary among mutation sites and types. Here, we generated knockout mice deficient in Cyp4f39, the mouse ortholog of human CYP4F22, to investigate the effects of completely abrogating the function of the fatty acid ω-hydroxylase involved in acylceramide production on skin barrier formation. Cyp4f39 knockout mice died within 8 hours of birth. Large increases in transepidermal water loss and penetration of a dye from outside the body were observed, indicating severe skin barrier dysfunction. Histologic analyses of the epidermis revealed impairment of lipid lamella formation, accumulation of corneodesmosomes in the stratum corneum, and persistence of periderm. In addition, lipid analyses by mass spectrometry showed almost complete loss of acylceramide and its precursor ω-hydroxy ceramide. In conclusion, our findings provide clues to the molecular mechanisms of skin barrier abnormalities and the pathogenesis of ichthyosis caused by Cyp4f39 and CYP4F22 by association.


Subject(s)
Ceramides/biosynthesis , Cytochrome P450 Family 4/metabolism , Epidermal Cells/pathology , Epidermis/pathology , Ichthyosis/pathology , Animals , Cytochrome P450 Family 4/genetics , Desmosomes/pathology , Disease Models, Animal , Epidermal Cells/cytology , Female , Humans , Ichthyosis/diagnosis , Ichthyosis/genetics , Male , Mice , Mice, Knockout , Permeability , Severity of Illness Index
12.
Front Immunol ; 10: 2883, 2019.
Article in English | MEDLINE | ID: mdl-31867019

ABSTRACT

Desmosomes reinforce cohesion of epithelial cells at the interface between adjacent cells. They include the cadherin-type adhesion molecules desmoglein 1 (Dsg1) and Dsg3. Pemphigus vulgaris (PV) is an autoimmune disease in which circulating autoantibodies (PV-IgG) targeting Dsg1 and 3 cause characteristic epidermal blister formation. It has been shown that PV-IgG binding induced activation of kinases such as ERK and PKC, and inhibition of these signaling pathways prevented loss of cell cohesion in cell cultures. However, the role of Erk and PKC in blister formation and regulation of desmosome ultrastructure in human skin are unknown. Accordingly, we assessed the role of PKC and ERK signaling pathways in blister formation and regulation of desmosome ultrastructure in human epidermis. Here we performed electron microscopy analyses using human skin explants injected with PV-IgG together with inhibitors for PKC or ERK signaling. Inhibition of PKC was not effective to prevent suprabasal blister formation or ultrastructural alterations of desmosomes. In contrast, inhibition of ERK signaling significantly ameliorated blister formation and decrease in the number of desmosomes whereas shortening and splitting of desmosomes and keratin filament insertion were not different from samples treated with PV-IgG alone. However, apical desmosomes between basal and suprabasal cells remained unaltered when ERK signaling was inhibited. Therefore, our results show that inhibition of ERK but not PKC signaling appears to be effective to ameliorate blistering and alterations of desmosome ultrastructure triggered by PV-IgG in human skin.


Subject(s)
Desmosomes/immunology , Epidermis/immunology , MAP Kinase Signaling System/immunology , Pemphigus/immunology , Protein Kinase C/immunology , Desmosomes/pathology , Epidermis/pathology , Humans , Pemphigus/pathology , Pemphigus/therapy
13.
Int J Mol Sci ; 20(16)2019 Aug 16.
Article in English | MEDLINE | ID: mdl-31426283

ABSTRACT

Arrhythmogenic cardiomyopathy (ACM) is an inherited heart disease characterized by sudden death in young people and featured by fibro-adipose myocardium replacement, malignant arrhythmias, and heart failure. To date, no etiological therapies are available. Mutations in desmosomal genes cause abnormal mechanical coupling, trigger pro-apoptotic signaling pathways, and induce fibro-adipose replacement. Here, we discuss the hypothesis that the ACM causative mechanism involves a defect in the expression and/or activity of the cardiac Ca2+ handling machinery, focusing on the available data supporting this hypothesis. The Ca2+ toolkit is heavily remodeled in cardiomyocytes derived from a mouse model of ACM defective of the desmosomal protein plakophilin-2. Furthermore, ACM-related mutations were found in genes encoding for proteins involved in excitation‒contraction coupling, e.g., type 2 ryanodine receptor and phospholamban. As a consequence, the sarcoplasmic reticulum becomes more eager to release Ca2+, thereby inducing delayed afterdepolarizations and impairing cardiac contractility. These data are supported by preliminary observations from patient induced pluripotent stem-cell-derived cardiomyocytes. Assessing the involvement of Ca2+ signaling in the pathogenesis of ACM could be beneficial in the treatment of this life-threatening disease.


Subject(s)
Arrhythmias, Cardiac/pathology , Calcium/metabolism , Cardiomyopathies/pathology , Desmosomes/pathology , Myocytes, Cardiac/pathology , Animals , Arrhythmias, Cardiac/metabolism , Calcium Signaling , Cardiomyopathies/metabolism , Desmosomes/metabolism , Humans , Myocytes, Cardiac/metabolism , Plakophilins/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism
14.
Biochim Biophys Acta Mol Basis Dis ; 1865(11): 165520, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31381994

ABSTRACT

Epidermal desquamation involves a finely-tuned proteolytic cascade ensuring the regulated cleavage of desmosomes that releases old stratum corneum outermost layers. Although the roles of desmosomes in normal physiology are well-established, their putative involvement in cancer remains unexplored. The KLK5 protease is thought of having fundamental roles in epidermal proteolysis and homeostasis, and its aberrant activity has been linked to skin pathologies. We found that deletion of Klk5 results in significantly higher numbers of lengthier desmosomes and enhanced skin strength. Klk5-/- mice retained normal skin barrier function and are resistant to chemically-induced skin tumorigenesis. The resistance to tumorigenesis was not due to inhibition of inflammation, and on the contrary, absence of Klk5 increased the TPA-induced inflammatory skin response. We found that increased desmosomes and reduced proteolysis prevent oncogenic signaling by capturing ß-catenin into the cytoplasm and facilitate epidermal keratinocyte apoptosis, thus, inhibiting tumor initiation. We highlight that the skin ultrastructure affects early neoplastic transformation by modulating intracellular signaling and suggest that tissue reinforcement provides a novel mode of tumor suppression.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Kallikreins/genetics , Skin Neoplasms/genetics , Animals , Carcinogenesis/pathology , Desmosomes/genetics , Desmosomes/pathology , Gene Deletion , Humans , Male , Mice , Mice, Inbred C57BL , Proteolysis , Skin/metabolism , Skin/pathology , Skin Neoplasms/pathology
15.
J Invest Dermatol ; 139(9): 1867-1875.e7, 2019 09.
Article in English | MEDLINE | ID: mdl-30905808

ABSTRACT

Atopic dermatitis is a common inflammatory skin disease caused by the interaction of genetic and environmental factors. By allelic copy number analysis at missense single-nucleotide polymorphisms on 26 genes with copy number variation, we identified a significant association between atopic dermatitis and human KPRP. Human KPRP expression, which was localized to the upper granular layer of epidermis, was significantly decreased in atopic dermatitis compared with normal skin. KPRP was histologically colocalized with loricrin and was mainly detected in cytoskeleton fractions of human keratinocytes. To further investigate the role of KPRP in skin, Kprp-knockout mice were generated. Heterozygous knockout (Kprp+/-) mice exhibited reduced KPRP expression to level a similar to that of human AD lesional skin. Kprp+/- mice showed abnormal desmosome structure and detachment of lower layers of the stratum corneum. Percutaneous inflammation by topical application of croton oil or oxazolone was enhanced, and epicutaneous immunization with ovalbumin induced a high level of IgE in Kprp+/- mice. Our study, started from allelic copy number analysis in human AD, identified the importance of KPRP, the decrease of which leads to barrier dysfunction.


Subject(s)
Cytoskeletal Proteins/genetics , Dermatitis, Atopic/genetics , Epidermis/pathology , Intracellular Signaling Peptides and Proteins/genetics , Keratinocytes/pathology , Proteins/genetics , Adjuvants, Immunologic/administration & dosage , Animals , Case-Control Studies , Croton Oil/immunology , Cytoskeletal Proteins/deficiency , DNA Copy Number Variations , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Desmosomes/pathology , Desmosomes/ultrastructure , Disease Models, Animal , Epidermis/drug effects , Epidermis/immunology , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Keratinocytes/drug effects , Keratinocytes/immunology , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Oxazolone/immunology , Proteins/metabolism , Water Loss, Insensible/genetics
16.
Cardiovasc Pathol ; 40: 2-6, 2019.
Article in English | MEDLINE | ID: mdl-30763825

ABSTRACT

Phospholamban (PLN) p.Arg14del cardiomyopathy is characterized by a distinct arrhythmogenic biventricular phenotype that can be predominantly left ventricular, right ventricular, or both. Our aim was to further elucidate distinct features of this cardiomyopathy with respect to the distribution of desmosomal proteins observed by immunofluorescence (IF) in comparison to desmosomal arrhythmogenic cardiomyopathy and co-existent genetic variants. We studied eight explanted heart specimens from PLN p.Arg14del mutation carriers. Macro- and microscopic examination revealed biventricular presence of fibrofatty replacement and interstitial fibrosis. Five out of 8 (63%) patients met consensus criteria for both arrhythmogenic right ventricular cardiomyopathy (ARVC) and dilated cardiomyopathy (DCM). In four cases, targeted next-generation sequencing revealed one additional pathogenic variant and six variants of unknown significance. IF showed diminished junction plakoglobin signal intensity at the intercalated disks in 4 (67%) out of 6 cases fulfilling ARVC criteria but normal intensity in both cases fulfilling only DCM criteria. Notably, the four cases with diminished junction plakoglobin were also those where an additional gene variant was detected. IF for two proteins recently investigated in desmosomal arrhythmogenic cardiomyopathy (ACM), synapse-associated protein 97 and glycogen synthase kinase-3 beta, showed a distinct distributional pattern in comparison to desmosomal ACM. In 7 (88%) out of 8 cases we observed both a strong synapse-associated protein 97 signal at the sarcomeres and no glycogen synthase kinase-3 beta translocation to the intercalated discs. Phospholamban p.Arg14del cardiomyopathy is characterized by a distinct molecular signature compared to desmosomal ACM, specifically a different desmosomal protein distribution. This study substantiates the idea that additional genetic variants play a role in the phenotypical heterogeneity.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia/genetics , Arrhythmogenic Right Ventricular Dysplasia/metabolism , Calcium-Binding Proteins/genetics , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Desmosomes/chemistry , Myocardium/chemistry , Sequence Deletion , Adipose Tissue/pathology , Adult , Aged , Arrhythmogenic Right Ventricular Dysplasia/pathology , Cardiomyopathy, Dilated/pathology , Desmosomes/pathology , Female , Fibrosis , Genetic Markers , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Myocardium/pathology , Phenotype , Prognosis , Registries , Risk Factors , Tight Junction Proteins/analysis
17.
J Cell Sci ; 132(5)2019 02 18.
Article in English | MEDLINE | ID: mdl-30659114

ABSTRACT

Arrhythmogenic cardiomyopathy (AC) is an incurable progressive disease that is linked to mutations in genes coding for components of desmosomal adhesions that are localized to the intercalated disc region, which electromechanically couples adjacent cardiomyocytes. To date, the underlying molecular dysfunctions are not well characterized. In two murine AC models, we find an upregulation of the skeletal muscle actin gene (Acta1), which is known to be a compensatory reaction to compromised heart function. Expression of this gene is elevated prior to visible morphological alterations and clinical symptoms, and persists throughout pathogenesis with an additional major rise during the chronic disease stage. We provide evidence that the increased Acta1 transcription is initiated through nuclear activation of the serum response transcription factor (SRF) by its transcriptional co-activator megakaryoblastic leukemia 1 protein (MKL1, also known as MRTFA). Our data further suggest that perturbed desmosomal adhesion causes Acta1 overexpression during the early stages of the disease, which is amplified by transforming growth factor ß (TGFß) release from fibrotic lesions and surrounding cardiomyocytes during later disease stages. These observations highlight a hitherto unknown molecular AC pathomechanism.


Subject(s)
Actins/genetics , Arrhythmogenic Right Ventricular Dysplasia/metabolism , Desmoglein 2/genetics , Desmosomes/metabolism , Muscle, Skeletal/metabolism , Mutation/genetics , Myocardium/pathology , Actins/metabolism , Animals , Arrhythmogenic Right Ventricular Dysplasia/genetics , Arrhythmogenic Right Ventricular Dysplasia/pathology , Cell Adhesion , Cells, Cultured , Desmoglein 2/metabolism , Desmosomes/pathology , Disease Models, Animal , Fibrosis , Humans , Mice , Mice, Mutant Strains , Myocardium/metabolism , Serum Response Factor/genetics , Serum Response Factor/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptional Activation , Transforming Growth Factor beta/metabolism , Up-Regulation
18.
J Invest Dermatol ; 139(2): 380-390, 2019 02.
Article in English | MEDLINE | ID: mdl-30321533

ABSTRACT

Investigation of genetic determinants of Mendelian skin disorders has substantially advanced understanding of epidermal biology. Here we show that mutations in PERP, encoding a crucial component of desmosomes, cause both dominant and recessive human keratoderma. Heterozygosity for a C-terminal truncation, which produces a protein that appears to be unstably incorporated into desmosomes, causes Olmsted syndrome with severe periorificial and palmoplantar keratoderma in multiple unrelated kindreds. Homozygosity for an N-terminal truncation ablates expression and causes widespread erythrokeratoderma, with expansion of epidermal differentiation markers. Both exhibit epidermal hyperproliferation, immature desmosomes lacking a dense midline observed via electron microscopy, and impaired intercellular adhesion upon mechanical stress. Localization of other desmosomal components appears normal, which is in contrast to other conditions caused by mutations in genes encoding desmosomal proteins. These discoveries highlight the essential role of PERP in human desmosomes and epidermal homeostasis and further expand the heterogeneous spectrum of inherited keratinization disorders.


Subject(s)
Desmosomes/pathology , Epidermis/pathology , Keratoderma, Palmoplantar/genetics , Membrane Proteins/genetics , Adult , Cell Adhesion/genetics , Child , Child, Preschool , Codon, Nonsense , DNA Mutational Analysis , Desmosomes/ultrastructure , Epidermis/ultrastructure , Exons/genetics , Female , Frameshift Mutation , Genes, Tumor Suppressor , Heterozygote , Homozygote , Humans , Keratoderma, Palmoplantar/pathology , Male , Membrane Proteins/metabolism , Microscopy, Electron , Young Adult
19.
Front Immunol ; 9: 1935, 2018.
Article in English | MEDLINE | ID: mdl-30233569

ABSTRACT

Pemphigus vulgaris (PV) is a potentially life-threatening autoimmune blistering disease which is associated with autoantibodies directed against two desmosomal proteins, desmoglein (Dsg) 3 and 1. Treatment of PV is rather challenging and relies on the long-term use of systemic corticosteroids and additional immunosuppressants. More recently, autoantibody-depleting therapies such as rituximab, high-dose intravenous immunoglobulins, and immunoadsorption were shown to be valuable treatment options in PV. Specific removal of pathogenic autoantibodies would further increase efficacy and usability of immunoadsorption. Here, we tested the capacity of our recently developed prototypic Dsg1- and Dsg3-specific adsorbers to remove circulating pathogenic autoantibodies from three different PV patients. The pathogenic potential of the Dsg3/1-depleted IgG fractions and the anti-Dsg3-specific IgG was explored in two different in vitro assays based on cultured human keratinocytes, the desmosome degradation assay and the dispase-based dissociation assay. In addition, the neonatal mouse model of PV was used. In both in vitro assays, no difference between the pathogenic effect of total PV IgG and anti-Dsg3-specific IgG was seen, while Dsg3/1-depleted and control IgG were not pathogenic. For the samples of all 3 PV patients, depletion of anti-Dsg3/1 IgG resulted in a complete loss of pathogenicity when injected into neonatal mice. In contrast, injection of anti-Dsg3-specific IgG, eluted from the column, induced gross blistering in the mice. Our data clearly show that anti-Dsg3-specific IgG alone is pathogenic in vitro and in vivo, whereas Dsg3/1-depletion results in a complete loss of pathogenicity. Furthermore, our data suggest that Dsg-specific adsorption may be a suitable therapeutic modality to efficiently reduce pathogenic autoantibodies in patients with severe PV.


Subject(s)
Antibodies, Anti-Idiotypic , Autoantibodies/immunology , Desmoglein 3/immunology , Desmosomes/immunology , Immunoglobulin G/immunology , Pemphigus/immunology , Adult , Aged , Animals , Antibodies, Anti-Idiotypic/immunology , Antibodies, Anti-Idiotypic/pharmacology , Autoantibodies/toxicity , Desmosomes/pathology , Female , Humans , Keratinocytes/immunology , Keratinocytes/pathology , Male , Mice , Pemphigus/pathology
20.
Front Immunol ; 9: 1190, 2018.
Article in English | MEDLINE | ID: mdl-29915578

ABSTRACT

Pemphigus vulgaris (PV) is a potentially life-threatening mucocutaneous autoimmune blistering disease. Patients develop non-healing erosions and blisters due to cell-cell detachment of keratinocytes (acantholysis), with subsequent suprabasal intraepidermal splitting. Identified almost 30 years ago, desmoglein-3 (Dsg3), a Ca2+-dependent cell adhesion molecule belonging to the cadherin family, has been considered the "primary" autoantigen in PV. Proteomic studies have identified numerous autoantibodies in patients with PV that have known roles in the physiology and cell adhesion of keratinocytes. Antibodies to these autoantibodies include desmocollins 1 and 3, several muscarinic and nicotinic acetylcholine receptor subtypes, mitochondrial proteins, human leukocyte antigen molecules, thyroid peroxidase, and hSPCA1-the Ca2+/Mn2+-ATPase encoded by ATP2C1, which is mutated in Hailey-Hailey disease. Several studies have identified direct pathogenic roles of these proteins, or synergistic roles when combined with Dsg3. We review the role of these direct and indirect mechanisms of non-desmoglein autoantibodies in the pathogenesis of PV.


Subject(s)
Autoantibodies/metabolism , Epitopes/immunology , Keratinocytes/physiology , Pemphigus, Benign Familial/immunology , Pemphigus/immunology , Animals , Autoantibodies/immunology , Desmoglein 3/immunology , Desmosomes/metabolism , Desmosomes/pathology , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...