1
|
Schmitt T, Huber J, Pircher J, Schmidt E, Waschke J. The impact of signaling pathways on the desmosome ultrastructure in pemphigus. Front Immunol 2025; 15:1497241. [PMID: 39882246 PMCID: PMC11774707 DOI: 10.3389/fimmu.2024.1497241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction The autoantibody-driven disease pemphigus vulgaris (PV) impairs desmosome adhesion in the epidermis. In desmosomes, the pemphigus autoantigens desmoglein 1 (Dsg1) and Dsg3 link adjacent cells. Dsgs are clustered by plaque proteins and linked to the keratin cytoskeleton by desmoplakin (Dp). The aim of this study was to identify the impact of several PV-related signaling pathways on desmosome ultrastructure. Methods STED microscopy, Dispase-based dissociation assay. Results As observed using STED microscopy, pemphigus autoantibodies (PV-IgG) reduced desmosome number, decreased desmosome size, increased plaque distance and thickness and caused loss of adhesion. Decreased desmosome number, increased plaque distance and thickness and loss of adhesion correlate with features found for newly assembled immature desmosomes, observed after Ca2+ depletion and repletion. This was paralleled by plaque asymmetry, keratin filament retraction and fragmentation of Dsg1 and Dsg3 immunostaining. Inhibition of each individual signaling pathway investigated here prevented the loss of adhesion and ameliorated keratin retraction. In addition, inhibition of p38MAPK or PLC completely rescued all parameters of desmosomes ultrastructure and increased desmosome number under basal conditions. In contrast, inhibition of MEK1/2 was only partially protective for desmosome size and plaque thickness, whereas inhibition of Src or increase of cAMP decreased desmosome size but increased the desmosome number even in the presence of PV-IgG. Discussion Alterations of the desmosomal plaque ultrastructure are closely related to loss of adhesion and regulated differently by signaling pathways involved in pemphigus pathogenesis. This insight may allow identification of novel treatment options targeting specific steps of desmosome turn-over in the future.
Collapse
Affiliation(s)
- Thomas Schmitt
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilan-Universität (LMU) Munich, München, Germany
| | - Julia Huber
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilan-Universität (LMU) Munich, München, Germany
| | - Julia Pircher
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilan-Universität (LMU) Munich, München, Germany
| | - Enno Schmidt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilan-Universität (LMU) Munich, München, Germany
| |
Collapse
|
2
|
Moghaddam AO, Jin X, Zhai H, Safa BT, Seiffert-Sinha K, Leiker M, Rosenbohm J, Meng F, Sinha AA, Yang R. Data-Driven Image Analysis to Determine Antibody-Induced Dissociation of Cell-Cell Adhesion and Antibody Pathogenicity in Pemphigus Vulgaris. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.09.617446. [PMID: 39416220 PMCID: PMC11482924 DOI: 10.1101/2024.10.09.617446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Pemphigus vulgaris (PV) is a blistering autoimmune disease that affects the skin and mucous membranes. The precise mechanisms by which PV antibodies induce a complete loss of cohesion of keratinocytes are not fully understood. But it is accepted that the process starts with antibody binding to desmosomal targets which leads to its disassembly and subsequent structural changes to cell-cell adhesions. In vitro immunofluorescence imaging of desmosome molecules has been used to characterize this initial phase, often qualitatively. However, there remains an untapped potential of image analysis in providing us more in-depth knowledge regarding ultrastructural changes after antibody binding. Currently, there is no such effort to establish a quantitative framework from immunofluorescence images in PV pathology. We take on this effort here in a comprehensive study to examine the effects of antibodies on key adhesion molecules and the cytoskeletal network, aiming to establish a correlation of ultrastructural changes in cell-cell adhesion with antibody pathogenicity. Specifically, we introduced a data-driven approach to quantitatively evaluate perturbations in adhesion molecules, including desmoglein 3, E-cadherin, as well as the cytoskeleton, following antibody treatment. We identify distinct immunofluorescence imaging signatures that mark the impact of antibody binding on the remodeling of the adhesion molecules and introduce a pathogenicity score to compare the relative effects of different antibodies. From this analysis, we showed that the biophysical response of keratinocytes to distinct PV associated antibodies is highly specific, allowing for accurate prediction of their pathogenicity. For instance, the high pathogenicity scores of the PVIgG and AK23 antibodies show strong agreement with their reported PV pathology. Our data-driven approach offers a more detailed framework for the action of autoantibodies in pemphigus and has the potential to pave the way for the development of effective novel diagnostic methods and therapeutic strategies.
Collapse
Affiliation(s)
- Amir Ostadi Moghaddam
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824
- Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
| | - Xiaowei Jin
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
| | - Haiwei Zhai
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
| | - Bahareh Tajvidi Safa
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824
- Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
| | | | - Merced Leiker
- Department of Dermatology, University at Buffalo, Buffalo, NY 14203
| | - Jordan Rosenbohm
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
| | | | - Animesh A. Sinha
- Department of Dermatology, University at Buffalo, Buffalo, NY 14203
| | - Ruiguo Yang
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824
- Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
| |
Collapse
|
3
|
Abstract
Desmosomes are relatives of ancient cadherin-based junctions, which emerged late in evolution to ensure the structural integrity of vertebrate tissues by coupling the intermediate filament cytoskeleton to cell-cell junctions. Their ability to dynamically counter the contractile forces generated by actin-associated adherens junctions is particularly important in tissues under high mechanical stress, such as the skin and heart. Much more than the simple cellular 'spot welds' depicted in textbooks, desmosomes are in fact dynamic structures that can sense and respond to changes in their mechanical environment and external stressors like ultraviolet light and pathogens. These environmental signals are transmitted intracellularly via desmosome-dependent mechanochemical pathways that drive the physiological processes of morphogenesis and differentiation. This Cell Science at a Glance article and the accompanying poster review desmosome structure and assembly, highlight recent insights into how desmosomes integrate chemical and mechanical signaling in the epidermis, and discuss desmosomes as targets in human disease.
Collapse
Affiliation(s)
- Abbey L. Perl
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jenny L. Pokorny
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kathleen J. Green
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
4
|
Xu T, Herkens L, Jia T, Klinkhammer BM, Kant S, Krusche CA, Buhl EM, Hayat S, Floege J, Strnad P, Kramann R, Djudjaj S, Boor P. The role of desmoglein-2 in kidney disease. Kidney Int 2024; 105:1035-1048. [PMID: 38395410 DOI: 10.1016/j.kint.2024.01.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/07/2023] [Accepted: 01/09/2024] [Indexed: 02/25/2024]
Abstract
Desmosomes are multi-protein cell-cell adhesion structures supporting cell stability and mechanical stress resilience of tissues, best described in skin and heart. The kidney is exposed to various mechanical stimuli and stress, yet little is known about kidney desmosomes. In healthy kidneys, we found desmosomal proteins located at the apical-junctional complex in tubular epithelial cells. In four different animal models and patient biopsies with various kidney diseases, desmosomal components were significantly upregulated and partly miss-localized outside of the apical-junctional complexes along the whole lateral tubular epithelial cell membrane. The most upregulated component was desmoglein-2 (Dsg2). Mice with constitutive tubular epithelial cell-specific deletion of Dsg2 developed normally, and other desmosomal components were not altered in these mice. When challenged with different types of tubular epithelial cell injury (unilateral ureteral obstruction, ischemia-reperfusion, and 2,8-dihydroxyadenine crystal nephropathy), we found increased tubular epithelial cell apoptosis, proliferation, tubular atrophy, and inflammation compared to wild-type mice in all models and time points. In vitro, silencing DSG2 via siRNA weakened cell-cell adhesion in HK-2 cells and increased cell death. Thus, our data show a prominent upregulation of desmosomal components in tubular cells across species and diseases and suggest a protective role of Dsg2 against various injurious stimuli.
Collapse
Affiliation(s)
- Tong Xu
- Institute of Pathology, RWTH Aachen University, Aachen, Germany; Department of Urology, the First Affiliated Hospital of Airforce Medical University, Xi'an, China
| | - Lea Herkens
- Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Ting Jia
- Institute of Pathology, RWTH Aachen University, Aachen, Germany; Department of Nephrology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | | | - Sebastian Kant
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Aachen, Germany
| | - Claudia A Krusche
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Aachen, Germany
| | - Eva M Buhl
- Electron Microscopy Facility, RWTH Aachen University, Aachen, Germany
| | - Sikander Hayat
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Jürgen Floege
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Pavel Strnad
- Department of Medicine III, Gastroenterology, Metabolic Diseases and Intensive Care, RWTH Aachen University, Aachen, Germany
| | - Rafael Kramann
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany; Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany; Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Sonja Djudjaj
- Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University, Aachen, Germany; Electron Microscopy Facility, RWTH Aachen University, Aachen, Germany; Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
5
|
Wang Y, Mang X, Li D, Wang Z, Chen Y, Cai Z, Tan F. Cold atmospheric plasma sensitizes head and neck cancer to chemotherapy and immune checkpoint blockade therapy. Redox Biol 2024; 69:102991. [PMID: 38103343 PMCID: PMC10764269 DOI: 10.1016/j.redox.2023.102991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023] Open
Abstract
Head and neck cancer (HNC) is the seventh most prevalent cancer globally, often characterized by chemo-resistance and immunosuppression, which significantly hampers treatment efficacy. Cold atmospheric plasma (CAP) has recently emerged as a promising adjuvant oncotherapy with substantial potential and advantages. In this study, Piezobrush® PZ2, a handheld CAP unit based on the piezoelectric direct discharge technology, was used to generate and deliver non-thermal plasma. We aimed to investigate the effects of CAPPZ2 on various types of HNC cells and elucidate the underlying mechanisms. In addition, we endeavored to examine the efficacy of combining CAPPZ2 with chemotherapy drugs (i.e., cisplatin) or immune checkpoint blockade (ICB, i.e., PD1 antibody) in HNC treatment. Firstly, the results demonstrated that CAPPZ2 exerted anti-neoplastic functions through inhibiting cell proliferation, migration and invasion, and promoting apoptosis and autophagy. Secondly, using transcriptomic sequencing, Western blotting, and quantitative real-time PCR, the mechanisms underlying CAPPZ2 treatment in vitro was presumed to be a multitargeted blockade of major cancer survival pathways, such as redox balance, glycolysis, and PI3K/AKT/mTOR/HIF-1α signaling. Lastly, combinatorial thearpy containing CAPPZ2 and cisplatin or PD-1 antibody significantly suppressed tumor growth and prolonged recipient survival in vivo. Collectively, the synergistic effects of CAPPZ2 and cisplatin or PD-1 antibody could serve as a promising solution to enhance head and neck tumor elimination.
Collapse
Affiliation(s)
- Yanhong Wang
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200432, China
| | - Xinyu Mang
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Danni Li
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200432, China
| | - Zhao Wang
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200432, China
| | - Yiliang Chen
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Zhenyu Cai
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Fei Tan
- Department of ORL-HNS, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200432, China; The Royal College of Surgeons in Ireland, Dublin, Ireland; The Royal College of Surgeons of England, London, UK.
| |
Collapse
|
6
|
Wan H, Teh MT, Mastroianni G, Ahmad US. Comparative Transcriptome Analysis Identifies Desmoglein-3 as a Potential Oncogene in Oral Cancer Cells. Cells 2023; 12:2710. [PMID: 38067138 PMCID: PMC10705960 DOI: 10.3390/cells12232710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/14/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
The role of desmoglein-3 (DSG3) in oncogenesis is unclear. This study aimed to uncover molecular mechanisms through comparative transcriptome analysis in oral cancer cells, defining potential key genes and associated biological processes related to DSG3 expression. Four mRNA libraries of oral squamous carcinoma H413 cell lines were sequenced, and 599 candidate genes exhibited differential expression between DSG3-overexpressing and matched control lines, with 12 genes highly significantly differentially expressed, including 9 upregulated and 3 downregulated. Genes with known implications in cancer, such as MMP-13, KRT84, OLFM4, GJA1, AMOT and ADAMTS1, were strongly linked to DSG3 overexpression. Gene ontology analysis indicated that the DSG3-associated candidate gene products participate in crucial cellular processes such as junction assembly, focal adhesion, extracellular matrix formation, intermediate filament organisation and keratinocyte differentiation. Validation of RNA-Seq was performed through RT-qPCR, Western blotting and immunofluorescence analyses. Furthermore, using transmission electron microscopy, we meticulously examined desmosome morphology and revealed a slightly immature desmosome structure in DSG3-overexpressing cells compared to controls. No changes in desmosome frequency and diameter were observed between the two conditions. This study underscores intricate and multifaceted alterations associated with DSG3 in oral squamous carcinoma cells, implying a potential oncogenic role of this gene in biological processes that enable cell communication, motility and survival.
Collapse
Affiliation(s)
- Hong Wan
- Center for Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Muy-Teck Teh
- Center for Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Giulia Mastroianni
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London E1 4NS, UK
| | - Usama Sharif Ahmad
- Center for Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|
7
|
Schmitt T, Hudemann C, Moztarzadeh S, Hertl M, Tikkanen R, Waschke J. Dsg3 epitope-specific signalling in pemphigus. Front Immunol 2023; 14:1163066. [PMID: 37143675 PMCID: PMC10151755 DOI: 10.3389/fimmu.2023.1163066] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
Introduction Pemphigus is an autoantibody driven disease that impairs the barrier function of the skin and mucosa by disrupting desmosomes and thereby impeding cellular cohesion. It is known that the different clinical phenotypes of pemphigus vulgaris (PV) and pemphigus foliaceus (PF) are dependent on the autoantibody profile and target antigens that, amongst others, are primarily desmoglein (Dsg)1 and/or Dsg3 for PV and Dsg1 for PF. However, it was reported that autoantibodiesagainst different epitopes of Dsg1 and Dsg3 can be pathogenic or not. The underlying mechanisms are very complex and involve both direct inhibition of Dsg interactions and downstream signalling. The aim of this study was to find out whether there is target-epitope-specific Dsg3 signalling by comparing the effects of the two pathogenic murine IgGs, 2G4 and AK23. Methods Dispase-based dissociation assay, Western Blot analysis, Stimulated emission depletion microscopy, Fura-based Ca2+ flux measurements, Rho/Rac G-Protein-linked immunosorbent assay, Enzyme-linked immunosorbent assay. Results The IgGs are directed against the EC5 and EC1 domain of Dsg3, respectively. The data show that 2G4 was less effective in causing loss of cell adhesion, compared to AK23. STED imaging revealed that both autoantibodies had similar effects on keratin retraction and reduction of desmosome number whereas only AK23 induced Dsg3 depletion. Moreover, both antibodies induced phosphorylation of p38MAPK and Akt whereas Src was phosphorylated upon treatment with AK23 only. Interestingly, Src and Akt activation were p38MAPK-dependent. All pathogenic effects were rescued by p38MAPK inhibition and AK23-mediated effects were also ameliorated by Src inhibition. Discussion The results give first insights into pemphigus autoantibody-induced Dsg3 epitope-specific signalling which is involved in pathogenic events such as Dsg3 depletion.
Collapse
Affiliation(s)
- Thomas Schmitt
- Instiute of Anatomy, Faculty of Medicine, Chair of Vegetative Anatomy, Ludwig-Maximilian -Universität (LMU) Munich, München, Germany
| | - Christoph Hudemann
- Department of Dermatology and Allergology, Philipps-University Marburg, Marburg, Germany
| | - Sina Moztarzadeh
- Instiute of Anatomy, Faculty of Medicine, Chair of Vegetative Anatomy, Ludwig-Maximilian -Universität (LMU) Munich, München, Germany
| | - Michael Hertl
- Department of Dermatology and Allergology, Philipps-University Marburg, Marburg, Germany
| | - Ritva Tikkanen
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University Giessen, Giessen, Germany
| | - Jens Waschke
- Instiute of Anatomy, Faculty of Medicine, Chair of Vegetative Anatomy, Ludwig-Maximilian -Universität (LMU) Munich, München, Germany
| |
Collapse
|
8
|
Fuchs M, Radeva MY, Spindler V, Vielmuth F, Kugelmann D, Waschke J. Cytoskeletal anchorage of different Dsg3 pools revealed by combination of hybrid STED/SMFS-AFM. Cell Mol Life Sci 2023; 80:25. [PMID: 36602635 PMCID: PMC9816259 DOI: 10.1007/s00018-022-04681-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023]
Abstract
Desmoglein 3 (Dsg3) is a desmosomal cadherin mediating cell adhesion within desmosomes and is the antigen of the autoimmune blistering skin disease pemphigus vulgaris. Therefore, understanding of the complex desmosome turnover process is of high biomedical relevance. Recently, super resolution microscopy was used to characterize desmosome composition and turnover. However, studies were limited because adhesion measurements on living cells were not possible in parallel. Before desmosomal cadherins are incorporated into nascent desmosomes, they are not bound to intermediate filaments but were suggested to be associated with the actin cytoskeleton. However, direct proof that adhesion of a pool of desmosomal cadherins is dependent on actin is missing. Here, we applied single-molecule force spectroscopy measurements with the novel single molecule hybrid-technique STED/SMFS-AFM to investigate the cytoskeletal anchorage of Dsg3 on living keratinocytes for the first time. By application of pharmacological agents we discriminated two different Dsg3 pools, only one of which is anchored to actin filaments. We applied the actin polymerization inhibitor Latrunculin B to modify the actin cytoskeleton and the PKCα activator PMA to modulate intermediate filament anchorage. On the cellular surface Dsg3 adhesion was actin-dependent. In contrast, at cell-cell contacts, Dsg3 adhesion was independent from actin but rather is regulated by PKC which is well established to control desmosome turn-over via intermediate filament anchorage. Taken together, using the novel STED/SMFS-AFM technique, we demonstrated the existence of two Dsg3 pools with different cytoskeletal anchorage mechanisms.
Collapse
Affiliation(s)
- Michael Fuchs
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Mariya Y Radeva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Volker Spindler
- Department of Biomedicine and Institute of Anatomy, University of Basel, Basel, Switzerland
| | - Franziska Vielmuth
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Daniela Kugelmann
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany.
| |
Collapse
|
9
|
Akhouayri L, Ostano P, Mello-Grand M, Gregnanin I, Crivelli F, Laurora S, Liscia D, Leone F, Santoro A, Mulè A, Guarino D, Maggiore C, Carlino A, Magno S, Scatolini M, Di Leone A, Masetti R, Chiorino G. Identification of a minimum number of genes to predict triple-negative breast cancer subgroups from gene expression profiles. Hum Genomics 2022; 16:70. [PMID: 36536459 PMCID: PMC9764480 DOI: 10.1186/s40246-022-00436-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/21/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a very heterogeneous disease. Several gene expression and mutation profiling approaches were used to classify it, and all converged to the identification of distinct molecular subtypes, with some overlapping across different approaches. However, a standardised tool to routinely classify TNBC in the clinics and guide personalised treatment is lacking. We aimed at defining a specific gene signature for each of the six TNBC subtypes proposed by Lehman et al. in 2011 (basal-like 1 (BL1); basal-like 2 (BL2); mesenchymal (M); immunomodulatory (IM); mesenchymal stem-like (MSL); and luminal androgen receptor (LAR)), to be able to accurately predict them. METHODS Lehman's TNBCtype subtyping tool was applied to RNA-sequencing data from 482 TNBC (GSE164458), and a minimal subtype-specific gene signature was defined by combining two class comparison techniques with seven attribute selection methods. Several machine learning algorithms for subtype prediction were used, and the best classifier was applied on microarray data from 72 Italian TNBC and on the TNBC subset of the BRCA-TCGA data set. RESULTS We identified two signatures with the 120 and 81 top up- and downregulated genes that define the six TNBC subtypes, with prediction accuracy ranging from 88.6 to 89.4%, and even improving after removal of the least important genes. Network analysis was used to identify highly interconnected genes within each subgroup. Two druggable matrix metalloproteinases were found in the BL1 and BL2 subsets, and several druggable targets were complementary to androgen receptor or aromatase in the LAR subset. Several secondary drug-target interactions were found among the upregulated genes in the M, IM and MSL subsets. CONCLUSIONS Our study took full advantage of available TNBC data sets to stratify samples and genes into distinct subtypes, according to gene expression profiles. The development of a data mining approach to acquire a large amount of information from several data sets has allowed us to identify a well-determined minimal number of genes that may help in the recognition of TNBC subtypes. These genes, most of which have been previously found to be associated with breast cancer, have the potential to become novel diagnostic markers and/or therapeutic targets for specific TNBC subsets.
Collapse
Affiliation(s)
- Laila Akhouayri
- Department of Biomedical Sciences, Genetics and Molecular Biology Laboratory, Faculty of Medicine and Pharmacy, Hassan II-Casablanca University, Casablanca, Morocco
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Paola Ostano
- Cancer Genomics Lab, Fondazione Edo ed Elvo Tempia, Biella, Italy
| | | | - Ilaria Gregnanin
- Cancer Genomics Lab, Fondazione Edo ed Elvo Tempia, Biella, Italy
| | - Francesca Crivelli
- Cancer Genomics Lab, Fondazione Edo ed Elvo Tempia, Biella, Italy
- Clinical Research Division, “Degli Infermi” Hospital, Ponderano, BI Italy
| | - Sara Laurora
- Molecular Oncology Lab, Fondazione Edo ed Elvo Tempia, Biella, Italy
| | - Daniele Liscia
- Pathology Department, “Degli Infermi” Hospital, Ponderano, BI Italy
| | - Francesco Leone
- Oncology Department, “Degli Infermi” Hospital, Ponderano, BI Italy
| | - Angela Santoro
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonino Mulè
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | - Claudia Maggiore
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Angela Carlino
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Stefano Magno
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Maria Scatolini
- Molecular Oncology Lab, Fondazione Edo ed Elvo Tempia, Biella, Italy
| | - Alba Di Leone
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Riccardo Masetti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | |
Collapse
|
10
|
Youssef RM, Saleh MA, Korany MM, Nour ZA, El-Kalioby M. Assessment of tissue E-cadherin and its proteolytic serum fragment in pemphigus vulgaris before and after remission: A case-control study. J Cosmet Dermatol 2022; 21:6343-6350. [PMID: 35912419 DOI: 10.1111/jocd.15284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 07/15/2022] [Accepted: 07/28/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND E-cadherin is a classic cadherin that mediates keratinocyte adhesion. AIMS To assess the tissue expression of E-cadherin and its proteolytic serum fragment (soluble E-cadherin) in pemphigus vulgaris (PV) before and after clinical remission compared with controls. PATIENTS Thirty-seven PV patients and thirty controls were enrolled. Pemphigus disease area index (PDAI) was calculated for patients at baseline and after remission. Punch biopsy specimens were taken from patients before, and after remission, and from controls for assessment of tissue E-cadherin by immunofluorescence. Similarly, serum samples were collected for assessment of serum soluble E-cadherin by ELISA. RESULTS Presence, intensity, and mean intensity of tissue E-cadherin were significantly reduced in PV patients before treatment compared with controls (p < 0.001). Detected E-cadherin showed mainly a basal and suprabasal distribution with cell surface and a cytoplasmic expression. Serum E-cadherin was significantly higher in patients before treatment compared with controls (p = 0.006). With remission, tissue E-cadherin presence, intensity, mean intensity, and serum E-cadherin showed statistically significant improvement (p = 0.003, <0.001, <0.001, and 0.003 respectively). Tissue E-cadherin presence and serum E-cadherin level reached values equivalent to the controls (p = 0.49 and 0.44, respectively). CONCLUSIONS Disruption of tissue E-cadherin and upregulation of serum soluble E-cadherin can contribute to the pathogenesis of PV. Clinical remission of PV is associated with normalization of tissue and serum E-cadherin.
Collapse
Affiliation(s)
| | - Marwah Adly Saleh
- Department of Dermatology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mona Mostafa Korany
- Department of Dermatology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Zeinab A Nour
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mona El-Kalioby
- Department of Dermatology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
11
|
Mizuta K, Matsubara T, Goto A, Addison WN, Nakatomi M, Matsuo K, Tada-Shigeyama Y, Yaginuma T, Honda H, Yoshioka I, Kokabu S. Plectin promotes tumor formation by B16 mouse melanoma cells via regulation of Rous sarcoma oncogene activity. BMC Cancer 2022; 22:936. [PMID: 36038818 PMCID: PMC9426213 DOI: 10.1186/s12885-022-10033-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 08/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Melanoma is a malignant tumor characterized by high proliferation and aggressive metastasis. To address the molecular mechanisms of the proto-oncogene, Rous sarcoma oncogene (Src), which is highly activated and promotes cell proliferation, migration, adhesion, and metastasis in melanoma. Plectin, a cytoskeletal protein, has recently been identified as a Src-binding protein that regulates Src activity in osteoclasts. Plectin is a candidate biomarker of certain tumors because of its high expression and the target of anti-tumor reagents such as ruthenium pyridinecarbothioamide. The molecular mechanisms by which plectin affects melanoma is still unclear. In this study, we examined the role of plectin in melanoma tumor formation. METHODS We used CRISPR/Cas9 gene editing to knock-out plectin in B16 mouse melanoma cells. Protein levels of plectin and Src activity were examined by western blotting analysis. In vivo tumor formation was assessed by subcutaneous injection of B16 cells into nude mice and histological analysis performed after 2 weeks by Hematoxylin-Eosin (H&E) staining. Cell proliferation was evaluated by direct cell count, cell counting kit-8 assays, cyclin D1 mRNA expression and Ki-67 immunostaining. Cell aggregation and adhesion were examined by spheroid formation, dispase-based dissociation assay and cell adhesion assays. RESULTS In in vivo tumor formation assays, depletion of plectin resulted in low-density tumors with large intercellular spaces. In vitro experiments revealed that plectin-deficient B16 cells exhibit reduced cell proliferation and reduced cell-to-cell adhesion. Since Src activity is reduced in plectin-deficient melanomas, we examined the relationship between plectin and Src signaling. Src overexpression in plectin knockout B16 cells rescued cell proliferation and improved cell-to-cell adhesion and cell to extracellular matrix adhesion. CONCLUSION These results suggest that plectin plays critical roles in tumor formation by promoting cell proliferation and cell-to-cell adhesion through Src signaling activity in melanoma cells.
Collapse
Affiliation(s)
- Kana Mizuta
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Japan.,Division of Oral Medicine, Department of Science of Physical Function, Kyushu Dental University, Kitakyushu, Japan
| | - Takuma Matsubara
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Japan.
| | - Akino Goto
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Japan
| | - William N Addison
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Japan
| | - Mitsushiro Nakatomi
- Department of Human, Information and Life Sciences, School of Health Sciences, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kou Matsuo
- Division of Oral Pathology, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Japan
| | - Yukiyo Tada-Shigeyama
- Division of Dental Anesthesiology, Department of Science of Physical Function, Kyushu Dental University, Kitakyushu, Japan
| | - Tatsuki Yaginuma
- Division of Oral and Maxillofacial Surgery, Department of Science and Physical Function, Kyushu Dental University, Kitakyushu, Japan
| | - Hiromi Honda
- School of Oral Health Sciences, Kyushu Dental University, Kitakyushu, Japan
| | - Izumi Yoshioka
- Division of Oral Medicine, Department of Science of Physical Function, Kyushu Dental University, Kitakyushu, Japan
| | - Shoichiro Kokabu
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Japan.
| |
Collapse
|
12
|
Schmitt T, Pircher J, Steinert L, Meier K, Ghoreschi K, Vielmuth F, Kugelmann D, Waschke J. Dsg1 and Dsg3 Composition of Desmosomes Across Human Epidermis and Alterations in Pemphigus Vulgaris Patient Skin. Front Immunol 2022; 13:884241. [PMID: 35711465 PMCID: PMC9196036 DOI: 10.3389/fimmu.2022.884241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
Desmosomes are important epidermal adhesion units and signalling hubs, which play an important role in pemphigus pathogenesis. Different expression patterns of the pemphigus autoantigens desmoglein (Dsg)1 and Dsg3 across different epidermal layers have been demonstrated. However, little is known about changes in desmosome composition in different epidermal layers or in patient skin. The aim of this study was thus to characterize desmosome composition in healthy and pemphigus skin using super-resolution microscopy. An increasing Dsg1/Dsg3 ratio from lower basal (BL) to uppermost granular layer (GL) was observed. Within BL desmosomes, Dsg1 and Dsg3 were more homogeneously distributed whereas superficial desmosomes mostly comprised one of the two molecules or domains containing either one but not both. Extradesmosomal, desmoplakin (Dp)-independent, co-localization of Dsg3 with plakoglobin (Pg) was found mostly in BL and extradesmosomal Dsg1 co-localization with Pg in all layers. In contrast, in the spinous layer (SL) most Dsg1 and Dsg3 staining was confined to desmosomes, as revealed by the co-localization with Dp. In pemphigus patient skin, Dsg1 and Dsg3 immunostaining was altered especially along blister edges. The number of desmosomes in patient skin was reduced significantly in basal and spinous layer keratinocytes with only few split desmosomes found. In addition, Dsg1-Pg co-localization at the apical BL and Dsg3-Pg co-localization in SL were significantly reduced in patients, suggesting that that extradesmosomal Dsg molecules were affected. These results support the hypothesis that pemphigus is a desmosome assembly disease and may help to explain histopathologic differences between pemphigus phenotypes.
Collapse
Affiliation(s)
- Thomas Schmitt
- Chair of Vegetative Anatomy, Instiute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-Universität München (LMU) Munich, München, Germany
| | - Julia Pircher
- Chair of Vegetative Anatomy, Instiute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-Universität München (LMU) Munich, München, Germany
| | - Letyfee Steinert
- Chair of Vegetative Anatomy, Instiute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-Universität München (LMU) Munich, München, Germany
| | - Katharina Meier
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berli, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berli, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Franziska Vielmuth
- Chair of Vegetative Anatomy, Instiute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-Universität München (LMU) Munich, München, Germany
| | - Daniela Kugelmann
- Chair of Vegetative Anatomy, Instiute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-Universität München (LMU) Munich, München, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Instiute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-Universität München (LMU) Munich, München, Germany
| |
Collapse
|
13
|
Brescacin A, Baig Z, Bhinder J, Lin S, Brar L, Cirillo N. What protein kinases are crucial for acantholysis and blister formation in pemphigus vulgaris? A systematic review. J Cell Physiol 2022; 237:2825-2837. [PMID: 35616233 PMCID: PMC9540544 DOI: 10.1002/jcp.30784] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 01/18/2023]
Abstract
Pemphigus vulgaris (PV) is a potentially fatal autoimmune blistering disease characterized by cell-cell detachment (or acantholysis) and blister formation. While the signaling mechanisms that associate with skin/mucosal blistering are being elucidated, specific treatment strategies targeting PV-specific pathomechanisms, particularly kinase signaling, have yet to be established. Hence, the aim of this review was to systematically evaluate molecules in the class of kinases that are essential for acantholysis and blister formation and are therefore candidates for targeted therapy. English articles from PubMed and Scopus databases were searched, and included in vitro, in vivo, and human studies that investigated the role of kinases in PV. We selected studies, extracted data and assessed risk of bias in duplicates and the results were reported according to the methodology outlined by the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA). The risk of bias assessment was performed on in vivo studies utilizing SYRCLE's risk of bias tool. Thirty-five studies were included that satisfied the pathogenicity criterion of kinases in PV, the vast majority being experimental models that used PV sera (n = 13) and PV-IgG (n = 22). Inhibition of kinase activity (p38MAPK, PKC, TK, c-Src, EGFR, ERK, mTOR, BTK, and CDK2) was achieved mostly by pharmacological means. Overall, we found substantial evidence that kinase inhibition reduced PV-associated phosphorylation events and keratinocyte disassociation, prevented acantholysis, and blocked blister formation. However, the scarce adherence to standardized reporting systems and the experimental protocols/models used did limit the internal and external validity of these studies. In summary, this systematic review highlighted the pathogenic intracellular events mediated by kinases in PV acantholysis and presented kinase signaling as a promising avenue for translational research. In particular, the molecules identified and discussed in this study represent potential candidates for the development of mechanism-based interventions in PV.
Collapse
Affiliation(s)
- Adriano Brescacin
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Carlton, Victoria, Australia
| | - Zunaira Baig
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Carlton, Victoria, Australia
| | - Jaspreet Bhinder
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Carlton, Victoria, Australia
| | - Sen Lin
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Carlton, Victoria, Australia
| | - Lovejot Brar
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Carlton, Victoria, Australia
| | - Nicola Cirillo
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Carlton, Victoria, Australia
| |
Collapse
|
14
|
Egu DT, Schmitt T, Waschke J. Mechanisms Causing Acantholysis in Pemphigus-Lessons from Human Skin. Front Immunol 2022; 13:884067. [PMID: 35720332 PMCID: PMC9205406 DOI: 10.3389/fimmu.2022.884067] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
Pemphigus vulgaris (PV) is an autoimmune bullous skin disease caused primarily by autoantibodies (PV-IgG) against the desmosomal adhesion proteins desmoglein (Dsg)1 and Dsg3. PV patient lesions are characterized by flaccid blisters and ultrastructurally by defined hallmarks including a reduction in desmosome number and size, formation of split desmosomes, as well as uncoupling of keratin filaments from desmosomes. The pathophysiology underlying the disease is known to involve several intracellular signaling pathways downstream of PV-IgG binding. Here, we summarize our studies in which we used transmission electron microscopy to characterize the roles of signaling pathways in the pathogenic effects of PV-IgG on desmosome ultrastructure in a human ex vivo skin model. Blister scores revealed inhibition of p38MAPK, ERK and PLC/Ca2+ to be protective in human epidermis. In contrast, inhibition of Src and PKC, which were shown to be protective in cell cultures and murine models, was not effective for human skin explants. The ultrastructural analysis revealed that for preventing skin blistering at least desmosome number (as modulated by ERK) or keratin filament insertion (as modulated by PLC/Ca2+) need to be ameliorated. Other pathways such as p38MAPK regulate desmosome number, size, and keratin insertion indicating that they control desmosome assembly and disassembly on different levels. Taken together, studies in human skin delineate target mechanisms for the treatment of pemphigus patients. In addition, ultrastructural analysis supports defining the specific role of a given signaling molecule in desmosome turnover at ultrastructural level.
Collapse
|
15
|
P-Cadherin Regulates Intestinal Epithelial Cell Migration and Mucosal Repair, but Is Dispensable for Colitis Associated Colon Cancer. Cells 2022; 11:cells11091467. [PMID: 35563773 PMCID: PMC9100778 DOI: 10.3390/cells11091467] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/20/2022] [Accepted: 04/23/2022] [Indexed: 12/16/2022] Open
Abstract
Recurrent chronic mucosal inflammation, a characteristic of inflammatory bowel diseases (IBD), perturbs the intestinal epithelial homeostasis resulting in formation of mucosal wounds and, in most severe cases, leads to colitis-associated colon cancer (CAC). The altered structure of epithelial cell-cell adhesions is a hallmark of intestinal inflammation contributing to epithelial injury, repair, and tumorigenesis. P-cadherin is an important adhesion protein, poorly expressed in normal intestinal epithelial cells (IEC) but upregulated in inflamed and injured mucosa. The goal of this study was to investigate the roles of P-cadherin in regulating intestinal inflammation and CAC. P-cadherin expression was markedly induced in the colonic epithelium of human IBD patients and CAC tissues. The roles of P-cadherin were investigated in P-cadherin null mice using dextran sulfate sodium (DSS)-induced colitis and an azoxymethane (AOM)/DSS induced CAC. Although P-cadherin knockout did not affect the severity of acute DSS colitis, P-cadherin null mice exhibited faster recovery after colitis. No significant differences in the number of colonic tumors were observed in P-cadherin null and control mice. Consistently, the CRISPR/Cas9-mediated knockout of P-cadherin in human IEC accelerated epithelial wound healing without affecting cell proliferation. The accelerated migration of P-cadherin depleted IEC was driven by activation of Src kinases, Rac1 GTPase and myosin II motors and was accompanied by transcriptional reprogramming of the cells. Our findings highlight P-cadherin as a negative regulator of IEC motility in vitro and mucosal repair in vivo. In contrast, this protein is dispensable for IEC proliferation and CAC development.
Collapse
|
16
|
Yang T, Jia L, Bian S, Chang X, Zhang Q, Tang Q, Zhu J, Yang Z, Feng Z. TROP2 Down-Regulated DSG2 to Promote Gastric Cancer Cell Invasion and Migration by EGFR/AKT and DSG2/PG/β-Catenin Pathways. Curr Cancer Drug Targets 2022; 22:691-702. [PMID: 35392784 DOI: 10.2174/1568009622666220407111013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/01/2021] [Accepted: 12/20/2021] [Indexed: 11/22/2022]
Abstract
AIMS Explore the specific mechanism of TROP2 in promoting cancer in gastric cancer, and provide a basis for the prevention and treatment of gastric cancer. Background Gastric cancer (GC) is the fourth most commonly found cancer and the second highest cause of cancer related death worldwide, TROP2 overexpression is closely related with many cancers including gastrointestinal tumors, DSG2 is an important protein in cell adhesion and its loss is related to cell migration. OBJECTIVE Explore the specific mechanism of TROP2 in promoting cancer in gastric cancer, and provide a basis for the prevention and treatment of gastric cancer. METHOD DSG2 was identified as an interacting protein of TROP2 in GC cells by co-immunoprecipitation and mass spectrometry. The regulated behavior of TROP2 on DSG2 expression was investigated with TROP2 over-expressure or knockdown. Cell-cell adhesion capacity medicated by DSG2 was evaluated by adhesion related assays. Electron microscope observation was utilized for accessing GC tumor desmosome assembly. Proteins in EGFR/AKT and DSG2/PG/β-catenin pathways were evaluated by western blotting. RESULT This study suggests that abundant expression of TROP2 in GC cells lessened DSG2 levels as well as desmosome adhesion, increased cell invasion, migration and promoted malignant progression through EGFR/AKT and DSG2/PG/β-catenin pathways. CONCLUSION TROP2 promotes gastric cancer cell invasion and migration by decreasing DSG2 expression through EGFR/AKT and DSG2/PG/β-catenin pathways.
Collapse
Affiliation(s)
- Tingting Yang
- Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China.,Department of Pathology, Nanjing Medical University, Nanjing 211166, China
| | - Lizhou Jia
- Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China
| | - Susu Bian
- Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China.,Department of Pathology, Nanjing Medical University, Nanjing 211166, China
| | - Xinxia Chang
- Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China
| | - Qian Zhang
- Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China
| | - Qi Tang
- Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China.,Department of Pathology, Nanjing Medical University, Nanjing 211166, China
| | - Jing Zhu
- Huadong Medical Institute of Biotechniques, Nanjing 210000, China
| | - Zhiping Yang
- Cancer Center, Bayannur Hospital, Bayannur, Inner Mongolia 015000, China
| | - Zhenqing Feng
- Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China.,Department of Pathology, Nanjing Medical University, Nanjing 211166, China.,Jiangsu Key Lab. of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
17
|
Yeruva S, Waschke J. Structure and regulation of desmosomes in intercalated discs: Lessons from epithelia. J Anat 2022; 242:81-90. [PMID: 35128661 PMCID: PMC9773171 DOI: 10.1111/joa.13634] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/22/2021] [Accepted: 01/10/2022] [Indexed: 12/25/2022] Open
Abstract
For electromechanical coupling of cardiomyocytes, intercalated discs (ICDs) are pivotal as highly specialized intercellular contact areas. ICD consists of adhesive contacts, such as desmosomes and adherens junctions (AJs) that are partially intermingled and thereby form an area composita to provide mechanical strength, as well as gap junctions (GJ) and sodium channels for excitation propagation. In contrast, in epithelia, mixed junctions with features of desmosomes and AJs are regarded as transitory primarily during the formation of desmosomes. The anatomy of desmosomes is defined by a typical ultrastructure with dense intracellular plaques anchoring the cadherin-type adhesion molecules to the intermediate filament cytoskeleton. Desmosomal diseases characterized by impaired adhesive and signalling functions of desmosomal contacts lead to arrhythmogenic cardiomyopathy when affecting cardiomyocytes and cause pemphigus when manifesting in keratinocytes or present as cardiocutaneous syndromes when both cell types are targeted by the disease, which underscores the high biomedical relevance of these cell contacts. Therefore, comparative analyses regarding the structure and regulation of desmosomal contacts in cardiomyocytes and epithelial cells are helpful to better understand disease pathogenesis. In this brief review, we describe the structural properties of ICD compared to epithelial desmosomes and suggest that mechanisms regulating adhesion may at least in part be comparable. Also, we discuss whether phenomena such as hyperadhesion or the bidirectional regulation of desmosomes to serve as signalling hubs in epithelial cells may also be relevant for ICD.
Collapse
Affiliation(s)
- Sunil Yeruva
- Ludwig‐Maximilian‐Universität München, Anatomische Anstalt, Lehrstuhl Anatomie I – Vegetative AnatomieMunichGermany
| | - Jens Waschke
- Ludwig‐Maximilian‐Universität München, Anatomische Anstalt, Lehrstuhl Anatomie I – Vegetative AnatomieMunichGermany
| |
Collapse
|
18
|
Kaur B, Kerbrat J, Kho J, Kaler M, Kanatsios S, Cirillo N. Mechanism-based therapeutic targets of pemphigus vulgaris: A scoping review of pathogenic molecular pathways. Exp Dermatol 2022; 31:154-171. [PMID: 34435386 DOI: 10.1111/exd.14453] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/20/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022]
Abstract
Pemphigus vulgaris (PV) is a potentially fatal autoimmune blistering disease characterised by cell-cell detachment or acantholysis. The mechanisms which follow antibody (Ab) binding and culminate in acantholytic changes and skin/mucosal blistering have not been fully clarified. Current treatment strategies are not specific to PV pathophysiology and although life-saving, harbour considerable side effects. We aimed to systematically assess the molecules amenable to targeted treatments that follow Ab binding and are associated with PV acantholysis. The resulting scoping review was conducted under PRISMA-ScR guidelines with clear inclusion and exclusion criteria and focused specifically on kinases, caspases, proteases, hydrolytic enzymes and other molecules of interest postulated to take part in the pathophysiology of PV. The review process resulted in the identification of 882 articles, of which 56 were eligible for qualitative synthesis. From the included articles, the majority (n = 42) used PV-IgG as the pathogenic agent, mainly via in vitro (n = 16) and in vivo (n = 10) models. Twenty-five molecules were found to play a pathogenic role in PV, including uPA, ADAM10, EGFR, Src, PKC, cdk2, ERK, PLC, calmodulin, NOS, p38MAPK and caspase-3. Selective inhibition of these molecules resulted in varying degrees of reduction in acantholysis and blistering. The pathogenic molecules identified in this review represent potential candidates for clinical translation.
Collapse
Affiliation(s)
- Bavleen Kaur
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| | - Jenna Kerbrat
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| | - Jia Kho
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| | - Manreet Kaler
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| | - Stefanos Kanatsios
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| | - Nicola Cirillo
- Melbourne Dental School, The University of Melbourne, Carlton, Victoria, Australia
| |
Collapse
|
19
|
Fuchs M, Kugelmann D, Schlegel N, Vielmuth F, Waschke J. Desmoglein 2 can undergo Ca2+-dependent interactions with both desmosomal and classical cadherins including E-cadherin and N-cadherin. Biophys J 2022; 121:1322-1335. [PMID: 35183520 PMCID: PMC9034291 DOI: 10.1016/j.bpj.2022.02.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/23/2021] [Accepted: 02/15/2022] [Indexed: 11/02/2022] Open
Abstract
Desmoglein (Dsg) 2 is a ubiquitously expressed desmosomal cadherin. Particularly, it is present in all cell types forming desmosomes, including epithelial cells and cardiac myocytes and is upregulated in the autoimmune skin disease pemphigus. Thus, we here characterized the binding properties of Dsg2 in more detail using atomic force microscopy (AFM). Dsg2 exhibits homophilic interactions and also heterophilic interactions with the desmosomal cadherin desmocollin (Dsc) 2, and further with the classical cadherins E-cadherin (E-Cad) and N-cadherin (N-Cad), which may be relevant for cross talk between desmosomes and adherens junctions in epithelia and cardiac myocytes. We found that all homo- and heterophilic interactions were Ca2+-dependent. All binding forces observed are in the same force range, i.e., 30 to 40 pN, except for the Dsg2/E-Cad unbinding force, which with 45 pN is significantly higher. To further characterize the nature of the interactions, we used tryptophan, a critical amino acid required for trans-interaction, and a tandem peptide (TP) designed to cross-link Dsg isoforms. TP was sufficient to prevent the tryptophan-induced loss of Dsg2 interaction with the desmosomal cadherins Dsg2 and Dsc2; however, not with the classical cadherins E-Cad and N-Cad, indicating that the interaction modes of Dsg2 with desmosomal and classical cadherins differ. TP rescued the tryptophan-induced loss of Dsg2 binding on living enterocytes, suggesting that interaction with desmosomal cadherins may be more relevant. In summary, the data suggest that the ubiquitous desmosomal cadherin Dsg2 enables the cross talk with adherens junctions by interacting with multiple binding partners with implications for proper adhesive function in healthy and diseased states.
Collapse
|
20
|
Yuan J, Jiang X, Lan H, Zhang X, Ding T, Yang F, Zeng D, Yong J, Niu B, Xiao S. Multi-Omics Analysis of the Therapeutic Value of MAL2 Based on Data Mining in Human Cancers. Front Cell Dev Biol 2022; 9:736649. [PMID: 35111745 PMCID: PMC8803135 DOI: 10.3389/fcell.2021.736649] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Recent studies have reported that T-cell differentiation protein 2 (MAL2) is an important regulator in cancers. Here, we downloaded data from multiple databases to analyze MAL2 expression and function in pan-cancers, especially in ovarian cancer (OC). Gene Expression Profiling Interactive Analysis (GEPIA) databases was used to examine MAL2 expression in 13 types of cancer. Kaplan–Meier plotter database was used to analyze the overall survival rate of MAL2 in pan-cancers. The Catalog of Somatic Mutations in Cancer (COSMIC), cBioPortal, and UCSC databases were used to examine MAL2 mutation in human cancers. Metascape, STRING, and GeneMANIA websites were used to explore MAL2 function in OC. Furthermore, ggplot2 package and ROC package were performed to analyze hub gene expression and undertake receiver operating characteristic (ROC) analysis. Drug sensitivity of MAL2 in OC was examined by the GSCALite database. In order to verify the results from databases above, real-time quantitative polymerase chain reaction (qRT-PCR) and western blotting were conducted to detect the expression of MAL2 in OC cells. CRISPR/Cas9 system was used to knockout the MAL2 gene in the OC cell lines HO8910 and OVCAR3, using specific guide RNA targeting the exons of MAL2. Then, we performed proliferation, colony formation, migration, and invasion assays to investigate the impact of MAL2 in OC cell lines in vivo and in vitro. Epithelial-mesenchymal transition (EMT)-associated biomarkers were significantly altered in vitro via western blotting and qRT-PCR. Taken together, we observed that MAL2 was remarkably dysregulated in multiple cancers and was related to patient overall survival (OS), mutation, and drug sensitivity. Furthermore, experimental results showed that MAL2 deletion negatively regulated the proliferation, migration, invasion, and EMT of OC, indicating that MAL2 is a novel oncogene that can activate EMT, significantly promote both the proliferation and migration of OC in vitro and in vivo, and provide new clues for treatment strategies.
Collapse
Affiliation(s)
- Jing Yuan
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyan Jiang
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Hua Lan
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyu Zhang
- School of Life Science and Technology, Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Tianyi Ding
- School of Life Science and Technology, Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Fan Yang
- School of Life Science and Technology, Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Da Zeng
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Jiahui Yong
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, China
| | - Beibei Niu
- Scientific Research Center, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Beibei Niu, ; Songshu Xiao,
| | - Songshu Xiao
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Beibei Niu, ; Songshu Xiao,
| |
Collapse
|
21
|
Schmitt T, Waschke J. Autoantibody-Specific Signalling in Pemphigus. Front Med (Lausanne) 2021; 8:701809. [PMID: 34434944 PMCID: PMC8381052 DOI: 10.3389/fmed.2021.701809] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/08/2021] [Indexed: 12/11/2022] Open
Abstract
Pemphigus is a severe autoimmune disease impairing barrier functions of epidermis and mucosa. Autoantibodies primarily target the desmosomal adhesion molecules desmoglein (Dsg) 1 and Dsg 3 and induce loss of desmosomal adhesion. Strikingly, autoantibody profiles in pemphigus correlate with clinical phenotypes. Mucosal-dominant pemphigus vulgaris (PV) is characterised by autoantibodies (PV-IgG) against Dsg3 whereas epidermal blistering in PV and pemphigus foliaceus (PF) is associated with autoantibodies against Dsg1. Therapy in pemphigus is evolving towards specific suppression of autoantibody formation and autoantibody depletion. Nevertheless, during the acute phase and relapses of the disease additional treatment options to stabilise desmosomes and thereby rescue keratinocyte adhesion would be beneficial. Therefore, the mechanisms by which autoantibodies interfere with adhesion of desmosomes need to be characterised in detail. Besides direct inhibition of Dsg adhesion, autoantibodies engage signalling pathways interfering with different steps of desmosome turn-over. With this respect, recent data indicate that autoantibodies induce separate signalling responses in keratinocytes via specific signalling complexes organised by Dsg1 and Dsg3 which transfer the signal of autoantibody binding into the cell. This hypothesis may also explain the different clinical pemphigus phenotypes.
Collapse
Affiliation(s)
- Thomas Schmitt
- Ludwig-Maximilian-Universität München, Anatomische Anstalt, Lehrstuhl Anatomie I - Vegetative Anatomie, Munich, Germany
| | - Jens Waschke
- Ludwig-Maximilian-Universität München, Anatomische Anstalt, Lehrstuhl Anatomie I - Vegetative Anatomie, Munich, Germany
| |
Collapse
|
22
|
Rehman A, Huang Y, Wan H. Evolving Mechanisms in the Pathophysiology of Pemphigus Vulgaris: A Review Emphasizing the Role of Desmoglein 3 in Regulating p53 and the Yes-Associated Protein. Life (Basel) 2021; 11:life11070621. [PMID: 34206820 PMCID: PMC8303937 DOI: 10.3390/life11070621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 01/28/2023] Open
Abstract
The immunobullous condition Pemphigus Vulgaris (PV) is caused by autoantibodies targeting the adhesion proteins of desmosomes, leading to blistering in the skin and mucosal membrane. There is still no cure to the disease apart from the use of corticosteroids and immunosuppressive agents. Despite numerous investigations, the pathological mechanisms of PV are still incompletely understood, though the etiology is thought to be multifactorial. Thus, further understanding of the molecular basis underlying this disease process is vital to develop targeted therapies. Ample studies have highlighted the role of Desmoglein-3 (DSG3) in the initiation of disease as DSG3 serves as a primary target of PV autoantibodies. DSG3 is a pivotal player in mediating outside-in signaling involved in cell junction remodeling, cell proliferation, differentiation, migration or apoptosis, thus validating its biological function in tissue integrity and homeostasis beyond desmosome adhesion. Recent studies have uncovered new activities of DSG3 in regulating p53 and the yes-associated protein (YAP), with the evidence of dysregulation of these pathways demonstrated in PV. The purpose of this review is to summarize the earlier and recent advances highlighting our recent findings related to PV pathogenesis that may pave the way for future research to develop novel specific therapies in curing this disease.
Collapse
Affiliation(s)
- Ambreen Rehman
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London, School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; (A.R.); (Y.H.)
- Department of Oral Diagnosis and Medicine, Dr Ishrat Ul Ebad Khan Institute of Oral Health Sciences, Dow University of Health Sciences, Karachi 74200, Pakistan
| | - Yunying Huang
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London, School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; (A.R.); (Y.H.)
| | - Hong Wan
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London, School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; (A.R.); (Y.H.)
- Correspondence:
| |
Collapse
|
23
|
Lowenthal BM, Sahoo D, Amin MB, Hansel DE. Urothelial Proliferation of Unknown Malignant Potential Involving the Bladder: Histopathologic Features and Risk of Progression in De Novo Cases and Cases With Prior Neoplasia. Arch Pathol Lab Med 2021; 144:853-862. [PMID: 31825667 DOI: 10.5858/arpa.2019-0005-oa] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2019] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Urothelial proliferation of unknown malignant potential (UPUMP) is a 2016 World Health Organization classifier that encompasses prior categories of flat and papillary urothelial hyperplasia. In addition, UPUMP occurs in settings of both de novo and prior bladder neoplasia. OBJECTIVE.— To identify UPUMP features associated with subsequent neoplastic development. DESIGN.— Sixty-eight patients were identified from the archives, including 26 patients with de novo and 42 patients with prior bladder neoplasia. Patient slides and clinical course were reviewed. RESULTS.— Patients with de novo UPUMP were detected through clinical findings (26/26; 100%), whereas surveillance cystoscopy primarily detected UPUMP in patients with prior neoplasia (29/42; 69%). Histopathologic criteria evaluated included urothelial hyperplasia, urothelial cytology, vascular ingrowth, denudation, inflammation, edema, and fibrosis. Mean clinical follow-up was 68.9 months in patients with de novo neoplasia and 69.5 months in patients with prior neoplasia. Subsequent neoplasia developed in 4 of 26 (15.4%) of patients with de novo UPUMP and was associated with cystoscopic papillary appearance (P = .02) or microscopic thin papillary ingrowths or papillations (P = .02; median time to progression, 4.1 months). Of 42 patients with prior neoplasia, 17 (40.5%) had subsequent neoplasia, significantly associated with an absence of prominent lamina propria edema (P < .001; median time to progression, 11.0 months). A higher rate of progression to high-grade disease was present in patients with a prior neoplasia versus those with de novo disease (58.9% versus 25%). CONCLUSIONS.— Urothelial proliferation of unknown malignant potential shows subsequent risk of neoplastic development of 17% in patients with de novo disease and 40% in patients with prior neoplasia. The greatest risk of progression is associated with early papillary formation.
Collapse
Affiliation(s)
- Brett M Lowenthal
- From the Department of Pathology (Drs Lowenthal and Hansel) and the Department of Computer Science and Engineering (Dr Sahoo), University of California, San Diego, La Jolla; and the Department of Pathology, University of Tennessee Health Sciences Center, Memphis (Dr Amin). Dr Lowenthal is now with Kaiser Permanente, San Diego, California. Dr Hansel is now with the Department of Pathology, Oregon Health & Science University, Portland
| | - Debashis Sahoo
- From the Department of Pathology (Drs Lowenthal and Hansel) and the Department of Computer Science and Engineering (Dr Sahoo), University of California, San Diego, La Jolla; and the Department of Pathology, University of Tennessee Health Sciences Center, Memphis (Dr Amin). Dr Lowenthal is now with Kaiser Permanente, San Diego, California. Dr Hansel is now with the Department of Pathology, Oregon Health & Science University, Portland
| | - Mahul B Amin
- From the Department of Pathology (Drs Lowenthal and Hansel) and the Department of Computer Science and Engineering (Dr Sahoo), University of California, San Diego, La Jolla; and the Department of Pathology, University of Tennessee Health Sciences Center, Memphis (Dr Amin). Dr Lowenthal is now with Kaiser Permanente, San Diego, California. Dr Hansel is now with the Department of Pathology, Oregon Health & Science University, Portland
| | - Donna E Hansel
- From the Department of Pathology (Drs Lowenthal and Hansel) and the Department of Computer Science and Engineering (Dr Sahoo), University of California, San Diego, La Jolla; and the Department of Pathology, University of Tennessee Health Sciences Center, Memphis (Dr Amin). Dr Lowenthal is now with Kaiser Permanente, San Diego, California. Dr Hansel is now with the Department of Pathology, Oregon Health & Science University, Portland
| |
Collapse
|
24
|
Xia H, Hu F, Pan L, Xu C, Huang H, Chen S, Ma H. FAM196B promotes proliferation and migration via regulating epithelial-mesenchymal transition in esophageal cancer. Cancer Biomark 2021; 31:39-46. [PMID: 33749638 DOI: 10.3233/cbm-203023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND EC (esophageal cancer) is a common cancer among people in the world. The molecular mechanism of FAM196B (family with sequence similarity 196 member B) in EC is still unclear. This article aimed to clarify the role of FAM196B in EC. METHODS The expression of FAM196B in EC tissues was detected using qRT-PCR. The prognosis of FAM196B in EC patients was determined by log-rank kaplan-Meier survival analysis and Cox regression analysis. Furthermore, shRNA was used to knockdown the expression of FAM196B in EC cell lines. MTT, wound healing assays and western blot were used to determine the role of FAM196B in EC cells. RESULTS In our research, we found that the expression of FAM196B was up-regulated in EC tissues. The increased expression of FAM196B was significantly correlated with differentiation, lymph node metastasis, stage, and poor survival. The proliferation and migration of EC cells were inhibited after FAM196B-shRNA transfection in vitro and vivo. The western blot result showed that FAM196B could regulate EMT. CONCLUSION These results suggested that FAM196B severs as an oncogene and promotes cell proliferation and migration in EC. In addition, FAM196B may be a potential therapeutic target for EC patients.
Collapse
Affiliation(s)
- Haifeng Xia
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Cardiothoracic Surgery, Suzhou Dushuhu Public Hospital, Suzhou, Jiangsu, China.,Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fang Hu
- Suzhou Kintor Pharmaceutical Limited, Suzhou, Jiangsu, China.,Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Liangbin Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chengcheng Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Haitao Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shaomu Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Haitao Ma
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
25
|
Wanuske M, Brantschen D, Schinner C, Stüdle C, Walter E, Hiermaier M, Vielmuth F, Waschke J, Spindler V. Clustering of desmosomal cadherins by desmoplakin is essential for cell-cell adhesion. Acta Physiol (Oxf) 2021; 231:e13609. [PMID: 33354837 DOI: 10.1111/apha.13609] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/19/2020] [Accepted: 12/20/2020] [Indexed: 12/14/2022]
Abstract
AIM Desmoplakin (Dp) is a crucial component of the desmosome, a supramolecular cell junction complex anchoring intermediate filaments. The mechanisms how Dp modulates cell-cell adhesion are only partially understood. Here, we studied the impact of Dp on the function of desmosomal adhesion molecules, desmosome turnover and intercellular adhesion. METHODS CRISPR/Cas9 was used for gene editing of human keratinocytes which were characterized by Western blot and immunostaining. Desmosomal ultrastructure and function were assessed by electron microscopy and cell adhesion assays. Single molecule binding properties and localization of desmosomal cadherins were studied by atomic force microscopy and super-resolution imaging. RESULTS Knockout (ko) of Dp impaired cell cohesion to drastically higher extents as ko of another desmosomal protein, plakoglobin (Pg). In contrast to Pg ko, desmosomes were completely absent in Dp ko. Binding properties of the desmosomal adhesion molecules desmocollin (Dsc) 3 and desmoglein (Dsg) 3 remained unaltered under loss of Dp. Dp was required for assembling desmosomal cadherins into large clusters, as Dsg2 and Dsc3, adhesion molecules primarily localized within desmosomes, were redistributed into small puncta in the cell membrane of Dp ko cells. Additional silencing of desmosomal cadherins in Dp ko did not further increase loss of intercellular adhesion. CONCLUSION Our data demonstrate that Dp is essential for desmosome formation but does not influence intercellular adhesion on the level of individual cadherin binding properties. Rather, macro-clustering of desmosomal adhesion molecules through Dp is crucial. These results may help to better understand severe diseases which are caused by Dp dysfunction.
Collapse
Affiliation(s)
- Marie‐Therès Wanuske
- Department of Biomedicine University of Basel Basel Switzerland
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | | | - Camilla Schinner
- Department of Biomedicine University of Basel Basel Switzerland
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | - Chiara Stüdle
- Department of Biomedicine University of Basel Basel Switzerland
| | - Elias Walter
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | - Matthias Hiermaier
- Department of Biomedicine University of Basel Basel Switzerland
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | - Franziska Vielmuth
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | - Jens Waschke
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| | - Volker Spindler
- Department of Biomedicine University of Basel Basel Switzerland
- Faculty of Medicine Ludwig‐Maximilians‐Universität Munich Munich Germany
| |
Collapse
|
26
|
Hiermaier M, Kliewe F, Schinner C, Stüdle C, Maly IP, Wanuske MT, Rötzer V, Endlich N, Vielmuth F, Waschke J, Spindler V. The Actin-Binding Protein α-Adducin Modulates Desmosomal Turnover and Plasticity. J Invest Dermatol 2020; 141:1219-1229.e11. [PMID: 33098828 DOI: 10.1016/j.jid.2020.09.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 01/01/2023]
Abstract
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.
Collapse
Affiliation(s)
- Matthias Hiermaier
- Department of Biomedicine, University of Basel, Basel, Switzerland; Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Felix Kliewe
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Camilla Schinner
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Chiara Stüdle
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - I Piotr Maly
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marie-Therès Wanuske
- Department of Biomedicine, University of Basel, Basel, Switzerland; Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Vera Rötzer
- Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Franziska Vielmuth
- Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Jens Waschke
- Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Volker Spindler
- Department of Biomedicine, University of Basel, Basel, Switzerland; Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany.
| |
Collapse
|
27
|
Schinner C, Olivares-Florez S, Schlipp A, Trenz S, Feinendegen M, Flaswinkel H, Kempf E, Egu DT, Yeruva S, Waschke J. The inotropic agent digitoxin strengthens desmosomal adhesion in cardiac myocytes in an ERK1/2-dependent manner. Basic Res Cardiol 2020; 115:46. [PMID: 32556797 PMCID: PMC7299919 DOI: 10.1007/s00395-020-0805-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 06/05/2020] [Indexed: 01/28/2023]
Abstract
Desmosomal proteins are components of the intercalated disc and mediate cardiac myocyte adhesion. Enhancement of cardiac myocyte cohesion, referred to as "positive adhesiotropy", was demonstrated to be a function of sympathetic signaling and to be relevant for a sufficient inotropic response. We used the inotropic agent digitoxin to investigate the link between inotropy and adhesiotropy. In contrast to wild-type hearts, digitoxin failed to enhance pulse pressure in perfused mice hearts lacking the desmosomal protein plakoglobin which was paralleled with abrogation of plaque thickening indicating that positive inotropic response requires intact desmosomal adhesion. Atomic force microscopy revealed that digitoxin increased the binding force of the adhesion molecule desmoglein-2 at cell-cell contact areas. This was paralleled by enhanced cardiac myocyte cohesion in both HL-1 cardiac myocytes and murine cardiac slices as determined by dissociation assays as well as by accumulation of desmosomal proteins at cell-cell contact areas. However, total protein levels or cytoskeletal anchorage were not affected. siRNA-mediated depletion of desmosomal proteins abrogated increase of cell cohesion demonstrating that intact desmosomal adhesion is required for positive adhesiotropy. Mechanistically, digitoxin caused activation of ERK1/2. In line with this, inhibition of ERK1/2 signaling abrogated the effects of digitoxin on cell-cell adhesion and desmosomal reorganization. These results show that the positive inotropic agent digitoxin enhances cardiac myocyte cohesion with reorganization of desmosomal proteins in an ERK1/2-dependent manner. Desmosomal adhesion seems to be important for a sufficient positive inotropic response of digitoxin treatment, which can be of medical relevance for the treatment of heart failure.
Collapse
Affiliation(s)
- Camilla Schinner
- Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Pettenkoferstraße 11, 80336, Munich, Germany
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Silvana Olivares-Florez
- Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Pettenkoferstraße 11, 80336, Munich, Germany
| | - Angela Schlipp
- Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Pettenkoferstraße 11, 80336, Munich, Germany
| | - Sebastian Trenz
- Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Pettenkoferstraße 11, 80336, Munich, Germany
| | - Manouk Feinendegen
- Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Pettenkoferstraße 11, 80336, Munich, Germany
| | - Heinrich Flaswinkel
- Department of Biology II, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Ellen Kempf
- Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Pettenkoferstraße 11, 80336, Munich, Germany
| | - Desalegn Tadesse Egu
- Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Pettenkoferstraße 11, 80336, Munich, Germany
| | - Sunil Yeruva
- Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Pettenkoferstraße 11, 80336, Munich, Germany
| | - Jens Waschke
- Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Pettenkoferstraße 11, 80336, Munich, Germany.
| |
Collapse
|
28
|
Xie HJ, Zhao J, Zhuo-Ma D, Zhan-Dui N, Er-Bu A, Tsering T. Inhibiting tumour metastasis by DQA modified paclitaxel plus ligustrazine micelles in treatment of non-small-cell lung cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 47:3465-3477. [PMID: 31432702 DOI: 10.1080/21691401.2019.1653900] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Lung cancer is a kind of malignant tumour characterized as uncontrolled cell growth in lung. These malignant cell growth can spread beyond the lung by process of metastasis into other tissues or parts of the body. In this study, we developed dequalinium (DQA) modified paclitaxel plus ligustrazine micelles to destroy vasculogenic mimicry (VM) channels and inhibit tumour metastasis. In vitro assays showed that the targeting micelles with centralized particle size distribution showed not only vigoroso cytotoxicity on A549 cells but also strong inhibition on VM channels and tumour metastasis. Mechanism studies indicated that the DQA modified paclitaxel plus ligustrazine micelles could down-regulate the expressions of VEGF, MMP2, TGF-β1 and E-cadherin in A549 cells. In vivo assays indicated that the targeting drug-loaded micelles could enhance the accumulation of chemotherapeutic drugs at tumour sites and exhibit strong tumour inhibitory activity with negligible toxicity. Hence, the DQA modified paclitaxel plus ligustrazine micelles developed in this study may provide a potential strategy for treatment of NSCLC.
Collapse
Affiliation(s)
- Hong-Jun Xie
- Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medicine College of Tibet University , Lhasa , China
| | - Jing Zhao
- Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medicine College of Tibet University , Lhasa , China
| | - DongZhi Zhuo-Ma
- Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medicine College of Tibet University , Lhasa , China
| | - NorBu Zhan-Dui
- Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medicine College of Tibet University , Lhasa , China
| | - Aga Er-Bu
- Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medicine College of Tibet University , Lhasa , China
| | - Tashi Tsering
- Traditional Tibetan Medical Research and Study Institute of Regional Traditional Tibetan Medical Hospital , Lhasa , China
| |
Collapse
|
29
|
Cheng J, Yang J, Xue K, Zhao Y, Zhao C, Li S, Wang Z. Desmoglein 3 Silencing Inhibits Inflammation and Goblet Cell Mucin Secretion in a Mouse Model of Chronic Rhinosinusitis via Disruption of the Wnt/β-Catenin Signaling Pathway. Inflammation 2020; 42:1370-1382. [PMID: 31028575 DOI: 10.1007/s10753-019-00998-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Chronic rhinosinusitis (CRS) is a common disease characterized by inflammation of the nose and paranasal sinuses lasting over 12 weeks. This study aims to evaluate the effect of desmoglein 3 (DSG3) on inflammatory response and goblet cell mucin secretion in a mouse model of CRS. The CRS-related differentially expressed genes and disease genes were screened using microarray-based gene expression analysis. Subsequently, CRS mouse models were established. The levels of pro-inflammatory factors TNF-α, IL-6, and IL-8 were measured by ELISA. In addition, loss-of-function experiment was conducted using siRNAs targeting DSG3 and β-catenin. The secretion of mucins MUC5B and MUC5AC in goblet cells was detected, and the apoptosis of goblet cells was assessed. The regulatory effect of DSG3 on the Wnt/β-catenin signaling pathway was analyzed by determining the mRNA and protein levels of DSG3, Wnt, β-catenin, and GSK3β. DSG3 was identified to be an upregulated gene in CRS, which was further documented in CRS mice models. Elevated inflammation and mucin production were noted in CRS mice models. Also, it was found that DSG3 or β-catenin silencing could decrease the levels of TNF-α, IL-6, and IL-8, and the positive rates of MUC5B and MUC5AC while enhancing goblet cell apoptosis. The Wnt/β-catenin signaling pathway was blocked by DSG3, evidenced by downregulated Wnt and β-catenin as well as upregulated GSK3β mRNA and protein levels. Overall, this study provides evidence that silencing DSG3 could inhibit the activation of the Wnt/β-catenin signaling pathway, thus alleviating CRS.
Collapse
Affiliation(s)
- Jinzhang Cheng
- Department of Otolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, Changchun, 130000, Jilin Province, People's Republic of China
| | - Jingpu Yang
- Department of Otolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, Changchun, 130000, Jilin Province, People's Republic of China
| | - Kai Xue
- Department of Otolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, Changchun, 130000, Jilin Province, People's Republic of China
| | - Yin Zhao
- Department of Otolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, Changchun, 130000, Jilin Province, People's Republic of China
| | - Chang Zhao
- Department of Otolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, Changchun, 130000, Jilin Province, People's Republic of China
| | - Song Li
- Department of Otolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, Changchun, 130000, Jilin Province, People's Republic of China
| | - Zonggui Wang
- Department of Otolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, Changchun, 130000, Jilin Province, People's Republic of China.
| |
Collapse
|
30
|
Uttagomol J, Ahmad US, Rehman A, Huang Y, Laly AC, Kang A, Soetaert J, Chance R, Teh MT, Connelly JT, Wan H. Evidence for the Desmosomal Cadherin Desmoglein-3 in Regulating YAP and Phospho-YAP in Keratinocyte Responses to Mechanical Forces. Int J Mol Sci 2019; 20:ijms20246221. [PMID: 31835537 PMCID: PMC6940936 DOI: 10.3390/ijms20246221] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/05/2019] [Accepted: 12/06/2019] [Indexed: 12/14/2022] Open
Abstract
Desmoglein 3 (Dsg3) plays a crucial role in cell-cell adhesion and tissue integrity. Increasing evidence suggests that Dsg3 acts as a regulator of cellular mechanotransduction, but little is known about its direct role in mechanical force transmission. The present study investigated the impact of cyclic strain and substrate stiffness on Dsg3 expression and its role in mechanotransduction in keratinocytes. A direct comparison was made with E-cadherin, a well-characterized mechanosensor. Exposure of oral and skin keratinocytes to equiaxial cyclic strain promoted changes in the expression and localization of junction assembly proteins. The knockdown of Dsg3 by siRNA blocked strain-induced junctional remodeling of E-cadherin and Myosin IIa. Importantly, the study demonstrated that Dsg3 regulates the expression and localization of yes-associated protein (YAP), a mechanosensory, and an effector of the Hippo pathway. Furthermore, we showed that Dsg3 formed a complex with phospho-YAP and sequestered it to the plasma membrane, while Dsg3 depletion had an impact on both YAP and phospho-YAP in their response to mechanical forces, increasing the sensitivity of keratinocytes to the strain or substrate rigidity-induced nuclear relocation of YAP and phospho-YAP. Plakophilin 1 (PKP1) seemed to be crucial in recruiting the complex containing Dsg3/phospho-YAP to the cell surface since its silencing affected Dsg3 junctional assembly with concomitant loss of phospho-YAP at the cell periphery. Finally, we demonstrated that this Dsg3/YAP pathway has an influence on the expression of YAP1 target genes and cell proliferation. Together, these findings provide evidence of a novel role for Dsg3 in keratinocyte mechanotransduction.
Collapse
Affiliation(s)
- Jutamas Uttagomol
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; (J.U.); (U.S.A.); (A.R.); (Y.H.); (A.K.); (R.C.); (M.-T.T.)
| | - Usama Sharif Ahmad
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; (J.U.); (U.S.A.); (A.R.); (Y.H.); (A.K.); (R.C.); (M.-T.T.)
| | - Ambreen Rehman
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; (J.U.); (U.S.A.); (A.R.); (Y.H.); (A.K.); (R.C.); (M.-T.T.)
| | - Yunying Huang
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; (J.U.); (U.S.A.); (A.R.); (Y.H.); (A.K.); (R.C.); (M.-T.T.)
| | - Ana C. Laly
- Centre for Cell Biology and Cutaneous Research, Blizard Institute; Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; (A.C.L.); (J.S.); (J.T.C.)
| | - Angray Kang
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; (J.U.); (U.S.A.); (A.R.); (Y.H.); (A.K.); (R.C.); (M.-T.T.)
| | - Jan Soetaert
- Centre for Cell Biology and Cutaneous Research, Blizard Institute; Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; (A.C.L.); (J.S.); (J.T.C.)
| | - Randy Chance
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; (J.U.); (U.S.A.); (A.R.); (Y.H.); (A.K.); (R.C.); (M.-T.T.)
| | - Muy-Teck Teh
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; (J.U.); (U.S.A.); (A.R.); (Y.H.); (A.K.); (R.C.); (M.-T.T.)
| | - John T. Connelly
- Centre for Cell Biology and Cutaneous Research, Blizard Institute; Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; (A.C.L.); (J.S.); (J.T.C.)
| | - Hong Wan
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; (J.U.); (U.S.A.); (A.R.); (Y.H.); (A.K.); (R.C.); (M.-T.T.)
- Correspondence: ; Tel.: +(44)-020-7882-7139; Fax: +(44)-020-7882-7137
| |
Collapse
|
31
|
Ocular surface involvement in pemphigus vulgaris: An interdisciplinary review. Ocul Surf 2019; 18:40-46. [PMID: 31614200 DOI: 10.1016/j.jtos.2019.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 09/21/2019] [Accepted: 09/25/2019] [Indexed: 11/23/2022]
Abstract
PURPOSE A review of the published literature on the history, pathogenesis, and treatment of pemphigus vulgaris (PV) and its ocular involvement. METHODS Literature searches were conducted in MEDLINE (Ovid), and google scholar for pemphigus vulgaris and ocular PV. Inclusion criteria were given to meta-analysis, case-controlled studies, and documented case reports. The data were examined and independently analyzed by more than two of the authors. RESULTS PV is a humoral autoimmune disease with a preponderance of IgG4 anti-desmoglein 3 antibodies. Upon antibody binding, there is an intracellular signaling mechanism that leads to blister formation. Ocular findings are seen in up to 16% of PV patients with conjunctivitis being the most common clinical presentation. New steroid-sparing agents have helped with the control of this deadly disease, and with better understanding of the pathogenesis of PV, other cytokine blockers currently available are promising steroid-sparing agents. CONCLUSIONS Ocular pemphigus can occasionally present prior to mucocutaneous findings. Recalcitrant conjunctivitis with conjunctival blisters should warrant a workup for systemic PV.
Collapse
|
32
|
E-Cadherin Is Important for Meibomian Gland Function as Revealed by a New Human ex Vivo Slice Culture Model. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1559-1568. [DOI: 10.1016/j.ajpath.2019.04.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/03/2019] [Accepted: 04/10/2019] [Indexed: 12/31/2022]
|
33
|
Kugelmann D, Rötzer V, Walter E, Egu DT, Fuchs MT, Vielmuth F, Vargas-Robles H, Schnoor M, Hertl M, Eming R, Rottner K, Schmidt A, Spindler V, Waschke J. Role of Src and Cortactin in Pemphigus Skin Blistering. Front Immunol 2019; 10:626. [PMID: 31024527 PMCID: PMC6461052 DOI: 10.3389/fimmu.2019.00626] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/08/2019] [Indexed: 02/06/2023] Open
Abstract
Autoantibodies against desmoglein (Dsg) 1 and Dsg3 primarily cause blister formation in the autoimmune disease pemphigus vulgaris (PV). Src was proposed to contribute to loss of keratinocyte cohesion. However, the role and underlying mechanisms are unclear and were studied here. In keratinocytes, cell cohesion in response to autoantibodies was reduced in Src-dependent manner by two patient-derived PV-IgG fractions as well as by AK23 but not by a third PV-IgG fraction, although Src was activated by all autoantibodies. Loss of cell cohesion was progredient in a timeframe of 24 h and AK23, similar to PV-IgG, interfered with reconstitution of cell cohesion after Ca2+-switch, indicating that the autoantibodies also interfered with desmosome assembly. Dsg3 co-localized along cell contacts and interacted with the Src substrate cortactin. In keratinocytes isolated from cortactin-deficient mice, cell adhesion was impaired and Src-mediated inhibition of AK23-induced loss of cell cohesion for 24 h was significantly reduced compared to wild-type (wt) cells. Similarly, AK23 impaired reconstitution of cell adhesion was Src-dependent only in the presence of cortactin. Likewise, Src inhibition significantly reduced AK23-induced skin blistering in wt but not cortactin-deficient mice. These data suggest that the Src-mediated long-term effects of AK23 on loss of cell cohesion and skin blistering are dependent on cortactin-mediated desmosome assembly. However, in human epidermis PV-IgG-induced skin blistering and ultrastructural alterations of desmosomes were not affected by Src inhibition, indicating that Src may not be critical for skin blistering in intact human skin, at least when high levels of autoantibodies targeting Dsg1 are present.
Collapse
Affiliation(s)
- Daniela Kugelmann
- Chair of Vegetative Anatomy, Institute of Anatomy, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Vera Rötzer
- Chair of Vegetative Anatomy, Institute of Anatomy, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Elias Walter
- Chair of Vegetative Anatomy, Institute of Anatomy, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Desalegn Tadesse Egu
- Chair of Vegetative Anatomy, Institute of Anatomy, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael Tobias Fuchs
- Chair of Vegetative Anatomy, Institute of Anatomy, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Franziska Vielmuth
- Chair of Vegetative Anatomy, Institute of Anatomy, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Michael Schnoor
- Department of Molecular Biomedicine, Cinvestav-IPN, Mexico City, Mexico
| | - Michael Hertl
- Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| | - Rüdiger Eming
- Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ansgar Schmidt
- Instiute of Pathology, Philipps-Universität Marburg, Marburg, Germany
| | - Volker Spindler
- Chair of Vegetative Anatomy, Institute of Anatomy, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
34
|
Choi YJ, Laclef C, Yang N, Andreu-Cervera A, Lewis J, Mao X, Li L, Snedecor ER, Takemaru KI, Qin C, Schneider-Maunoury S, Shroyer KR, Hannun YA, Koch PJ, Clark RA, Payne AS, Kowalczyk AP, Chen J. RPGRIP1L is required for stabilizing epidermal keratinocyte adhesion through regulating desmoglein endocytosis. PLoS Genet 2019; 15:e1007914. [PMID: 30689641 PMCID: PMC6366717 DOI: 10.1371/journal.pgen.1007914] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/07/2019] [Accepted: 12/24/2018] [Indexed: 02/01/2023] Open
Abstract
Cilia-related proteins are believed to be involved in a broad range of cellular processes. Retinitis pigmentosa GTPase regulator interacting protein 1-like (RPGRIP1L) is a ciliary protein required for ciliogenesis in many cell types, including epidermal keratinocytes. Here we report that RPGRIP1L is also involved in the maintenance of desmosomal junctions between keratinocytes. Genetically disrupting the Rpgrip1l gene in mice caused intraepidermal blistering, primarily between basal and suprabasal keratinocytes. This blistering phenotype was associated with aberrant expression patterns of desmosomal proteins, impaired desmosome ultrastructure, and compromised cell-cell adhesion in vivo and in vitro. We found that disrupting the RPGRIP1L gene in HaCaT cells, which do not form primary cilia, resulted in mislocalization of desmosomal proteins to the cytoplasm, suggesting a cilia-independent function of RPGRIP1L. Mechanistically, we found that RPGRIP1L regulates the endocytosis of desmogleins such that RPGRIP1L-knockdown not only induced spontaneous desmoglein endocytosis, as determined by AK23 labeling and biotinylation assays, but also exacerbated EGTA- or pemphigus vulgaris IgG-induced desmoglein endocytosis. Accordingly, inhibiting endocytosis with dynasore or sucrose rescued these desmosomal phenotypes. Biotinylation assays on cell surface proteins not only reinforced the role of RPGRIP1L in desmoglein endocytosis, but also suggested that RPGRIP1L may be more broadly involved in endocytosis. Thus, data obtained from this study advanced our understanding of the biological functions of RPGRIP1L by identifying its role in the cellular endocytic pathway.
Collapse
Affiliation(s)
- Yeon Ja Choi
- Department of Pathology, Stony Brook University, Stony Brook, NY, United States of America
| | - Christine Laclef
- Sorbonne Université, CNRS UMR7622, Inserm U1156, IBPS-Laboratoire de Biologie du Développement, Paris, France
| | - Ning Yang
- Department of Pathology, Stony Brook University, Stony Brook, NY, United States of America
| | - Abraham Andreu-Cervera
- Sorbonne Université, CNRS UMR7622, Inserm U1156, IBPS-Laboratoire de Biologie du Développement, Paris, France
| | - Joshua Lewis
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Xuming Mao
- Department of Dermatology, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Li Li
- Department of Dermatology, Peking Union Medical College Hospital, Beijing, China
| | - Elizabeth R Snedecor
- Department of Pathology, Stony Brook University, Stony Brook, NY, United States of America
| | - Ken-Ichi Takemaru
- Department of Pharmacology, Stony Brook University, Stony Brook, NY, United States of America
| | - Chuan Qin
- Institute of Laboratory Animal Science, Chinese Academy of Medical Science; and Comparative Medical Center, Peking Union Medical College, Beijing, China
| | - Sylvie Schneider-Maunoury
- Sorbonne Université, CNRS UMR7622, Inserm U1156, IBPS-Laboratoire de Biologie du Développement, Paris, France
| | - Kenneth R Shroyer
- Department of Pathology, Stony Brook University, Stony Brook, NY, United States of America
| | - Yusuf A Hannun
- Department of Medicine and Cancer Center, Stony Brook University, Stony Brook, NY, United States of America
| | - Peter J Koch
- Department of Dermatology and Center for Regenerative Medicine, University of Colorado Denver, Aurora, CO, United States of America
| | - Richard A Clark
- Department of Dermatology, Stony Brook University, Stony Brook, NY, United States of America
| | - Aimee S Payne
- Department of Dermatology, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Andrew P Kowalczyk
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Jiang Chen
- Department of Pathology, Stony Brook University, Stony Brook, NY, United States of America
- Institute of Laboratory Animal Science, Chinese Academy of Medical Science; and Comparative Medical Center, Peking Union Medical College, Beijing, China
- Department of Dermatology, Stony Brook University, Stony Brook, NY, United States of America
| |
Collapse
|
35
|
Waschke J. Desmogleins as signaling hubs regulating cell cohesion and tissue/organ function in skin and heart - EFEM lecture 2018. Ann Anat 2018; 226:96-100. [PMID: 30529571 DOI: 10.1016/j.aanat.2018.11.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 11/27/2018] [Indexed: 01/18/2023]
Abstract
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 β1-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.
Collapse
Affiliation(s)
- Jens Waschke
- Institute of Anatomy, Faculty of Medicine, LMU Munich, Pettenkoferstr. 11, 80336 Munich, Germany.
| |
Collapse
|
36
|
Ungewiß H, Rötzer V, Meir M, Fey C, Diefenbacher M, Schlegel N, Waschke J. Dsg2 via Src-mediated transactivation shapes EGFR signaling towards cell adhesion. Cell Mol Life Sci 2018; 75:4251-4268. [PMID: 29980799 PMCID: PMC11105603 DOI: 10.1007/s00018-018-2869-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 06/19/2018] [Accepted: 07/03/2018] [Indexed: 12/31/2022]
Abstract
Rapidly renewing epithelial tissues such as the intestinal epithelium require precise tuning of intercellular adhesion and proliferation to preserve barrier integrity. Here, we provide evidence that desmoglein 2 (Dsg2), an adhesion molecule of desmosomes, controls cell adhesion and proliferation via epidermal growth factor receptor (EGFR) signaling. Dsg2 is required for EGFR localization at intercellular junctions as well as for Src-mediated EGFR activation. Src binds to EGFR and is required for localization of EGFR and Dsg2 to cell-cell contacts. EGFR is critical for cell adhesion and barrier recovery. In line with this, Dsg2-deficient enterocytes display impaired barrier properties and increased cell proliferation. Mechanistically, Dsg2 directly interacts with EGFR and undergoes heterotypic-binding events on the surface of living enterocytes via its extracellular domain as revealed by atomic force microscopy. Thus, our study reveals a new mechanism by which Dsg2 via Src shapes EGFR function towards cell adhesion.
Collapse
Affiliation(s)
- Hanna Ungewiß
- Department I, Institute of Anatomy and Cell Biology, Ludwig Maximilians University Munich, Pettenkoferstr. 11, 80336, Munich, Germany
| | - Vera Rötzer
- Department I, Institute of Anatomy and Cell Biology, Ludwig Maximilians University Munich, Pettenkoferstr. 11, 80336, Munich, Germany
| | - Michael Meir
- Department of General, Visceral, Vascular and Paediatric Surgery, Julius-Maximilians-Universität, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Christina Fey
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070, Würzburg, Germany
| | - Markus Diefenbacher
- Department of Biochemistry and Molecular Biochemistry, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Nicolas Schlegel
- Department of General, Visceral, Vascular and Paediatric Surgery, Julius-Maximilians-Universität, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Jens Waschke
- Department I, Institute of Anatomy and Cell Biology, Ludwig Maximilians University Munich, Pettenkoferstr. 11, 80336, Munich, Germany.
| |
Collapse
|
37
|
Bhandari A, Shen Y, Sindan N, Xia E, Gautam B, Lv S, Zhang X. MAL2 promotes proliferation, migration, and invasion through regulating epithelial-mesenchymal transition in breast cancer cell lines. Biochem Biophys Res Commun 2018; 504:434-439. [PMID: 30195491 DOI: 10.1016/j.bbrc.2018.08.187] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 08/29/2018] [Accepted: 08/29/2018] [Indexed: 01/18/2023]
Abstract
BACKGROUND Breast cancer is one of the most common malignant tumors in women. However, the underlying molecular mechanisms of breast cancer are still far to clear. With the development of sequencing technology, we discovered that MAL2 is overexpressed in tumor tissues. But the major function of MAL2 in breast cancer has not to be well confirmed. MATERIALS AND METHODS We downloaded and analyzed the MAL2 expression in The Cancer Genome Atlas (TCGA) database. Real-time quantitative polymerase chain reaction (RT-qPCR) was conducted to detect the expression of MAL2 in 35 breast cancer patients. Then, we performed proliferation, colony formation, migration, invasion and western blot assays to investigate the role of MAL2 in breast cancer cell lines (MDA-MB-231 and BT-549). RESULTS In our research, we found that MAL2 is remarkably overexpressed in breast cancer tissues compared to adjacent non-cancer tissues by RT-qPCR (T: N = 5.28 ± 4.34:1.82 ± 1.11, P < 0.001) and high expression of MAL2 has worse overall survival in TCGA cohort (P = 0.0032). Knocked down MAL2 could decrease the ability of proliferation, migration, and invasion of breast cancer cell lines. Our Western Blot assay results investigated that MAL2 could regulate EMT. CONCLUSION In this study, we demonstrated the function of MAL2 in breast cancer cell lines and it might act as an oncogene in breast cancer.
Collapse
Affiliation(s)
- Adheesh Bhandari
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, PR China
| | - Yanyan Shen
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, PR China
| | - Namita Sindan
- Department of Reproductive Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, PR China
| | - Erjie Xia
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, PR China
| | - Bishnu Gautam
- School of International Studies, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, PR China
| | - Shixu Lv
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, PR China.
| | - Xiaohua Zhang
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, PR China.
| |
Collapse
|
38
|
Amber KT, Valdebran M, Grando SA. Non-Desmoglein Antibodies in Patients With Pemphigus Vulgaris. Front Immunol 2018; 9:1190. [PMID: 29915578 PMCID: PMC5994403 DOI: 10.3389/fimmu.2018.01190] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 05/14/2018] [Indexed: 12/14/2022] Open
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.
Collapse
Affiliation(s)
- Kyle T Amber
- Department of Dermatology, University of California Irvine, Irvine, CA, United States
| | - Manuel Valdebran
- Department of Dermatology, University of California Irvine, Irvine, CA, United States
| | - Sergei A Grando
- Department of Dermatology, University of California Irvine, Irvine, CA, United States.,Department of Dermatology, Institute for Immunology, University of California Irvine, Irvine, CA, United States.,Department of Biological Chemistry, Institute for Immunology, University of California Irvine, Irvine, CA, United States
| |
Collapse
|
39
|
Basu S, Chaudhary N, Shah S, Braggs C, Sawant A, Vaz S, Thorat R, Gupta S, Dalal SN. Plakophilin3 loss leads to an increase in lipocalin2 expression, which is required for tumour formation. Exp Cell Res 2018; 369:251-265. [PMID: 29803740 DOI: 10.1016/j.yexcr.2018.05.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 12/17/2022]
Abstract
An increase in tumour formation and metastasis are observed upon plakophilin3 (PKP3) loss. To identify pathways downstream of PKP3 loss that are required for increased tumour formation, a gene expression analysis was performed, which demonstrated that the expression of lipocalin2 (LCN2) was elevated upon PKP3 loss and this is consistent with expression data from human tumour samples suggesting that PKP3 loss correlates with an increase in LCN2 expression. PKP3 loss leads to an increase in invasion, tumour formation and metastasis and these phenotypes were dependent on the increase in LCN2 expression. The increased LCN2 expression was due to an increase in the activation of p38 MAPK in the HCT116 derived PKP3 knockdown clones as LCN2 expression decreased upon inhibition of p38 MAPK. The phosphorylated active form of p38 MAPK is translocated to the nucleus upon PKP3 loss and is dependent on complex formation between p38 MAPK and PKP3. WT PKP3 inhibits LCN2 reporter activity in PKP3 knockdown cells but a PKP3 mutant that fails to form a complex with p38 MAPK cannot suppress LCN2 promoter activity. Further, LCN2 expression is decreased upon loss of p38β, but not p38α, in the PKP3 knockdown cells. These results suggest that PKP3 loss leads to an increase in the nuclear translocation of p38 MAPK and p38β MAPK is required for the increase in LCN2 expression.
Collapse
Affiliation(s)
- Srikanta Basu
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Nazia Chaudhary
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Sanket Shah
- Epigenetics and Chromatin Biology Group, Gupta Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Carol Braggs
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Aakanksha Sawant
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Simone Vaz
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Rahul Thorat
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Sanjay Gupta
- Epigenetics and Chromatin Biology Group, Gupta Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Sorab N Dalal
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India.
| |
Collapse
|
40
|
Vielmuth F, Spindler V, Waschke J. Atomic Force Microscopy Provides New Mechanistic Insights into the Pathogenesis of Pemphigus. Front Immunol 2018; 9:485. [PMID: 29643851 PMCID: PMC5883869 DOI: 10.3389/fimmu.2018.00485] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/23/2018] [Indexed: 12/19/2022] Open
Abstract
Autoantibodies binding to the extracellular domains of desmoglein (Dsg) 3 and 1 are critical in the pathogenesis of pemphigus by mechanisms leading to impaired function of desmosomes and blister formation in the epidermis and mucous membranes. Desmosomes are highly organized protein complexes which provide strong intercellular adhesion. Desmosomal cadherins such as Dsgs, proteins of the cadherin superfamily which interact via their extracellular domains in Ca2+-dependent manner, are the transmembrane adhesion molecules clustered within desmosomes. Investigations on pemphigus cover a wide range of experimental approaches including biophysical methods. Especially atomic force microscopy (AFM) has recently been applied increasingly because it allows the analysis of native materials such as cultured cells and tissues under near-physiological conditions. AFM provides information about the mechanical properties of the sample together with detailed interaction analyses of adhesion molecules. With AFM, it was recently demonstrated that autoantibodies directly inhibit Dsg interactions on the surface of living keratinocytes, a phenomenon which has long been considered the main mechanism causing loss of cell cohesion in pemphigus. In addition, AFM allows to study how signaling pathways altered in pemphigus control binding properties of Dsgs. More general, AFM and other biophysical studies recently revealed the importance of keratin filaments for regulation of Dsg binding and keratinocyte mechanical properties. In this mini-review, we reevaluate AFM studies in pemphigus and keratinocyte research, recapitulate what is known about the interaction mechanisms of desmosomal cadherins and discuss the advantages and limitations of AFM in these regards.
Collapse
Affiliation(s)
| | | | - Jens Waschke
- Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
41
|
Vielmuth F, Walter E, Fuchs M, Radeva MY, Buechau F, Magin TM, Spindler V, Waschke J. Keratins Regulate p38MAPK-Dependent Desmoglein Binding Properties in Pemphigus. Front Immunol 2018; 9:528. [PMID: 29616033 PMCID: PMC5868517 DOI: 10.3389/fimmu.2018.00528] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/28/2018] [Indexed: 12/20/2022] Open
Abstract
Keratins are crucial for the anchorage of desmosomes. Severe alterations of keratin organization and detachment of filaments from the desmosomal plaque occur in the autoimmune dermatoses pemphigus vulgaris and pemphigus foliaceus (PF), which are mainly caused by autoantibodies against desmoglein (Dsg) 1 and 3. Keratin alterations are a structural hallmark in pemphigus pathogenesis and correlate with loss of intercellular adhesion. However, the significance for autoantibody-induced loss of intercellular adhesion is largely unknown. In wild-type (wt) murine keratinocytes, pemphigus autoantibodies induced keratin filament retraction. Under the same conditions, we used murine keratinocytes lacking all keratin filaments (KtyII k.o.) as a model system to dissect the role of keratins in pemphigus. KtyII k.o. cells show compromised intercellular adhesion without antibody (Ab) treatment, which was not impaired further by pathogenic pemphigus autoantibodies. Nevertheless, direct activation of p38MAPK via anisomycin further decreased intercellular adhesion indicating that cell cohesion was not completely abrogated in the absence of keratins. Direct inhibition of Dsg3, but not of Dsg1, interaction via pathogenic autoantibodies as revealed by atomic force microscopy was detectable in both cell lines demonstrating that keratins are not required for this phenomenon. However, PF-IgG shifted Dsg1-binding events from cell borders toward the free cell surface in wt cells. This led to a distribution pattern of Dsg1-binding events similar to KtyII k.o. cells under resting conditions. In keratin-deficient keratinocytes, PF-IgG impaired Dsg1-binding strength, which was not different from wt cells under resting conditions. In addition, pathogenic autoantibodies were capable of activating p38MAPK in both KtyII wt and k.o. cells, the latter of which already displayed robust p38MAPK activation under resting conditions. Since inhibition of p38MAPK blocked autoantibody-induced loss of intercellular adhesion in wt cells and restored baseline cell cohesion in keratin-deficient cells, we conclude that p38MAPK signaling is (i) critical for regulation of cell adhesion, (ii) regulated by keratins, and (iii) targets both keratin-dependent and -independent mechanisms.
Collapse
Affiliation(s)
- Franziska Vielmuth
- Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Elias Walter
- Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael Fuchs
- Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Mariya Y Radeva
- Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Fanny Buechau
- Division of Cell and Developmental Biology, Institute of Biology, Sächsische Inkubator für Klinische Translation (SIKT), University of Leipzig, Leipzig, Germany
| | - Thomas M Magin
- Division of Cell and Developmental Biology, Institute of Biology, Sächsische Inkubator für Klinische Translation (SIKT), University of Leipzig, Leipzig, Germany
| | - Volker Spindler
- Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jens Waschke
- Faculty of Medicine, Institute of Anatomy, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
42
|
Spindler V, Waschke J. Pemphigus-A Disease of Desmosome Dysfunction Caused by Multiple Mechanisms. Front Immunol 2018; 9:136. [PMID: 29449846 PMCID: PMC5799217 DOI: 10.3389/fimmu.2018.00136] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/16/2018] [Indexed: 02/01/2023] Open
Abstract
Pemphigus is a severe autoimmune-blistering disease of the skin and mucous membranes caused by autoantibodies reducing desmosomal adhesion between epithelial cells. Autoantibodies against the desmosomal cadherins desmogleins (Dsgs) 1 and 3 as well as desmocollin 3 were shown to be pathogenic, whereas the role of other antibodies is unclear. Dsg3 interactions can be directly reduced by specific autoantibodies. Autoantibodies also alter the activity of signaling pathways, some of which regulate cell cohesion under baseline conditions and alter the turnover of desmosomal components. These pathways include Ca2+, p38MAPK, PKC, Src, EGFR/Erk, and several others. In this review, we delineate the mechanisms relevant for pemphigus pathogenesis based on the histology and the ultrastructure of patients’ lesions. We then dissect the mechanisms which can explain the ultrastructural hallmarks detectable in pemphigus patient skin. Finally, we reevaluate the concept that the spectrum of mechanisms, which induce desmosome dysfunction upon binding of pemphigus autoantibodies, finally defines the clinical phenotype.
Collapse
Affiliation(s)
- Volker Spindler
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Jens Waschke
- Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| |
Collapse
|
43
|
Spindler V, Eming R, Schmidt E, Amagai M, Grando S, Jonkman MF, Kowalczyk AP, Müller EJ, Payne AS, Pincelli C, Sinha AA, Sprecher E, Zillikens D, Hertl M, Waschke J. Mechanisms Causing Loss of Keratinocyte Cohesion in Pemphigus. J Invest Dermatol 2017; 138:32-37. [PMID: 29037765 DOI: 10.1016/j.jid.2017.06.022] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 11/28/2022]
Abstract
The autoimmune blistering skin disease pemphigus is caused by IgG autoantibodies against desmosomal cadherins, but the precise mechanisms are in part a matter of controversial discussions. This review focuses on the currently existing models of the disease and highlights the relevance of desmoglein-specific versus nondesmoglein autoantibodies, the contribution of nonautoantibody factors, and the mechanisms leading to cell dissociation and blister formation in response to autoantibody binding. As the review brings together the majority of laboratories currently working on pemphigus pathogenesis, it aims to serve as a solid basis for further investigations for the entire field.
Collapse
Affiliation(s)
- Volker Spindler
- Institute of Anatomy and Cell Biology, Ludwig-Maximilians-Universität, Munich, Germany.
| | - Rüdiger Eming
- Department of Dermatology, University of Marburg, Marburg, Germany
| | - Enno Schmidt
- Department of Dermatology, University of Lübeck, Lübeck, Germany; Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Sergei Grando
- Institute for Immunology and Departments of Dermatology and Biological Chemistry, University of California, Irvine, California, USA
| | - Marcel F Jonkman
- Department of Dermatology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Andrew P Kowalczyk
- Departments of Cell Biology and Dermatology, Emory University, Atlanta, Georgia, USA
| | - Eliane J Müller
- Vetsuisse Faculty, Molecular Dermatology and Stem Cell Research, Institute of Animal Pathology, Bern, Switzerland; Vetsuisse Faculty, DermFocus, Bern, Switzerland; Department of Dermatology, University Hospital of Bern, Bern, Switzerland
| | - Aimee S Payne
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Carlo Pincelli
- Laboratory of Cutaneous Biology, University of Modena and Reggio Emilia, Modena, Italy
| | - Animesh A Sinha
- Department of Dermatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Eli Sprecher
- Department of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Detlef Zillikens
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Michael Hertl
- Department of Dermatology, University of Marburg, Marburg, Germany
| | - Jens Waschke
- Institute of Anatomy and Cell Biology, Ludwig-Maximilians-Universität, Munich, Germany.
| |
Collapse
|
44
|
Dudley JS, Murphy CR, Thompson MB, McAllan BM. Epithelial cadherin disassociates from the lateral plasma membrane of uterine epithelial cells throughout pregnancy in a marsupial. J Anat 2017; 231:359-365. [PMID: 28670836 PMCID: PMC5554831 DOI: 10.1111/joa.12648] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2017] [Indexed: 01/31/2023] Open
Abstract
The uterine luminal epithelium is the first site of contact between fetal and maternal tissues during therian pregnancy and must undergo specialised changes for implantation of the blastocyst to be successful. These changes, collectively termed the plasma membrane transformation (PMT), allow the blastocyst to attach to the uterine epithelium preceding the formation of a placenta. There are similarities in the morphological and molecular changes occurring in live-bearing eutherian species during the PMT studied so far. Similar cellular remodelling occurs in a marsupial species, the fat-tailed dunnart (Sminthopsis crassicaudata), despite the divergence of marsupials from eutherian mammals over 130 mya, which resulted in the evolution of distinct reproductive strategies. Adhesion molecules along the lateral plasma membrane of uterine epithelium provide a barrier to invasion by the embryo. We thus characterised the presence and change in distribution of epithelial cadherin (E-cadherin) in uterine epithelium from non-pregnant fat-tailed dunnarts and compared it to dunnarts in early-, mid- and late-stage pregnancy. E-cadherin staining is localised to the lateral plasma membrane in uterine epithelium from non-pregnant and early-stage pregnant dunnarts. The E-cadherin staining is cytoplasmic in epithelium from uteri of mid- and late-stage pregnant dunnarts. This loss of localised staining suggests that the adherens junction dissociates from the lateral plasma membrane, allowing for invasion between the epithelial cells by the blastocyst. As the changes during pregnancy to cadherin were similar in the laboratory rat with highly invasive (haemochorial) placentation, a live-bearing lizard species with non-invasive (epitheliochorial) placentation and a marsupial, the fat-tailed dunnart, which has invasive (endotheliochorial) placentation, we suggest that the molecular mechanisms allowing for successful pregnancy are conserved among mammals during the early stages of pregnancy regardless of placental invasiveness.
Collapse
Affiliation(s)
- Jessica S. Dudley
- School of Medical Sciences and Bosch InstituteUniversity of SydneySydneyNSWAustralia
| | - Christopher R. Murphy
- School of Medical Sciences and Bosch InstituteUniversity of SydneySydneyNSWAustralia
| | - Michael B. Thompson
- School of Life and Environmental ScienceUniversity of SydneySydneyNSWAustralia
| | - Bronwyn M. McAllan
- School of Medical Sciences and Bosch InstituteUniversity of SydneySydneyNSWAustralia
| |
Collapse
|
45
|
Desmoglein 2 regulates the intestinal epithelial barrier via p38 mitogen-activated protein kinase. Sci Rep 2017; 7:6329. [PMID: 28740231 PMCID: PMC5524837 DOI: 10.1038/s41598-017-06713-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 06/16/2017] [Indexed: 01/18/2023] Open
Abstract
Intestinal epithelial barrier properties are maintained by a junctional complex consisting of tight junctions (TJ), adherens junctions (AJ) and desmosomes. Desmoglein 2 (Dsg2), an adhesion molecule of desmosomes and the only Dsg isoform expressed in enterocytes, is required for epithelial barrier properties and may contribute to barrier defects in Crohn’s disease. Here, we identified extradesmosomal Dsg2 on the surface of polarized enterocytes by Triton extraction, confocal microscopy, SIM and STED. Atomic force microscopy (AFM) revealed Dsg2-specific binding events along the cell border on the surface of enterocytes with a mean unbinding force of around 30pN. Binding events were blocked by an inhibitory antibody targeting Dsg2 which under same conditions activated p38MAPK but did not reduce cell cohesion. In enterocytes deficient for Dsg2, p38MAPK activity was reduced and both barrier integrity and reformation were impaired. Dsc2 rescue did not restore p38MAPK activity indicating that Dsg2 is required. Accordingly, direct activation of p38MAPK in Dsg2-deficient cells enhanced barrier reformation demonstrating that Dsg2-mediated activation of p38MAPK is crucial for barrier function. Collectively, our data show that Dsg2, beside its adhesion function, regulates intestinal barrier function via p38MAPK signalling. This is in contrast to keratinocytes and points towards tissue-specific signalling functions of desmosomal cadherins.
Collapse
|
46
|
Schmidt E, Spindler V, Eming R, Amagai M, Antonicelli F, Baines JF, Belheouane M, Bernard P, Borradori L, Caproni M, Di Zenzo G, Grando S, Harman K, Jonkman MF, Koga H, Ludwig RJ, Kowalczyk AP, Müller EJ, Nishie W, Pas H, Payne AS, Sadik CD, Seppänen A, Setterfield J, Shimizu H, Sinha AA, Sprecher E, Sticherling M, Ujiie H, Zillikens D, Hertl M, Waschke J. Meeting Report of the Pathogenesis of Pemphigus and Pemphigoid Meeting in Munich, September 2016. J Invest Dermatol 2017; 137:1199-1203. [PMID: 28390814 DOI: 10.1016/j.jid.2017.01.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 01/05/2017] [Indexed: 02/07/2023]
Abstract
Autoimmune blistering diseases are a heterogeneous group of about a dozen complex disorders that are characterized by intraepidermal (pemphigus) and subepidermal blistering (pemphigoid diseases and dermatitis herpetiformis). The Pathogenesis of Pemphigus and Pemphigoid Meeting, organized by the Departments of Dermatology in Lübeck and Marburg and the Institute of Anatomy and Cell Biology, Munich, was held in September 2016 in Munich. The meeting brought together basic scientists and clinicians from all continents dedicating their work to autoimmune blistering diseases. Considerable advances have been made in describing incidences and prevalences of these diseases and linking comorbidities with autoantibody reactivities and clinical variants, for example, dipeptidyl peptidase-IV inhibitor-associated noninflammatory bullous pemphigoid. Although new entities are still being described, diagnosis of most autoimmune blistering diseases can now be achieved using standardized and widely available serological test systems. Various experimental mouse models of pemphigus and pemphigoid disease are increasingly being used to understand mechanisms of central and peripheral tolerance and to evaluate more specific treatment approaches for these disorders, such as molecules that target autoreactive T and B cells and anti-inflammatory mediators, that is, dimethyl fumarate, phosphodiesterase 4, and leukotriene B4 inhibitors in pemphigoid disorders, and chimeric antigen receptor T cells in pemphigus. Very recent experimental data about the immunopathology and the determinants of autoantibody formation and keratinocyte susceptibility in pemphigus were discussed. With regard to cellular mechanisms leading to the loss of cell-cell adhesion, new ideas were shared in the field of signal transduction. Major steps were taken to put the various partly contradictory and controversial findings about the effects of pemphigus autoantibodies and other inflammatory mediators into perspective and broaden our view of the complex pathophysiology of this disease. Finally, two investigator-initiated multicenter trials highlighted doxycycline and dapsone as valuable medications in the treatment of bullous pemphigoid.
Collapse
Affiliation(s)
- Enno Schmidt
- Department of Dermatology, University of Lübeck, Lübeck, Germany; Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany.
| | - Volker Spindler
- Institute of Anatomy and Cell Biology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Rüdiger Eming
- Department of Dermatology, Philipps University, Marburg, Germany
| | | | | | - John F Baines
- Max Planck Institute for Evolutionary Biology, Plön, and Institute for Experimental Medicine, University of Kiel, Kiel, Germany
| | - Meriem Belheouane
- Max Planck Institute for Evolutionary Biology, Plön, and Institute for Experimental Medicine, University of Kiel, Kiel, Germany
| | | | - Luca Borradori
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Marzia Caproni
- Department of Dermatology, University of Florence, Florence, Italy
| | - Giovanni Di Zenzo
- Molecular and Cell Biology Laboratory, Istituto Dermopatico dell'Immacolata (IDI)-IRCCS FLMM, Rome, Italy
| | - Sergei Grando
- Institute for Immunology and Departments of Dermatology and Biological Chemistry, University of California, Irvine, California, USA
| | - Karen Harman
- Department of Dermatology, University Hospitals of Leicester, Leicester, UK
| | - Marcel F Jonkman
- Department of Dermatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Hiroshi Koga
- Department of Dermatology, Kurume University School of Medicine, Kurume, Japan
| | - Ralf J Ludwig
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Andrew P Kowalczyk
- Departments of Cell Biology and Dermatology, Emory University, Atlanta, Georgia, USA
| | - Eliane J Müller
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Institute of Animal Pathology, University of Bern, Switzerland
| | - Wataru Nishie
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hendri Pas
- Department of Dermatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Aimee S Payne
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Allan Seppänen
- Department of Psychiatry, Helsinki University Hospital, Helsinki, Finland
| | - Jane Setterfield
- Mucosal and Salivary Biology, Dental Institute, King's College London & St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Hiroshi Shimizu
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Animesh A Sinha
- Department of Dermatology, Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, Buffalo, New York, USA
| | - Eli Sprecher
- Department of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | | | - Hideyuki Ujiie
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Detlef Zillikens
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Michael Hertl
- Department of Dermatology, Philipps University, Marburg, Germany
| | - Jens Waschke
- Institute of Anatomy and Cell Biology, Ludwig-Maximilians-Universität, Munich, Germany
| |
Collapse
|
47
|
Hütz K, Zeiler J, Sachs L, Ormanns S, Spindler V. Loss of desmoglein 2 promotes tumorigenic behavior in pancreatic cancer cells. Mol Carcinog 2017; 56:1884-1895. [PMID: 28277619 DOI: 10.1002/mc.22644] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 02/17/2017] [Accepted: 03/03/2017] [Indexed: 12/31/2022]
Abstract
The ability to maintain cell-cell adhesion is crucial for tissue integrity and organization. Accordingly, loss of cohesiveness plays a critical role in cancer invasion and metastasis. Desmosomes are cell junctions providing strong intercellular adhesive strength and dysregulation of desmosomal constituents contributes to cancer progression through altered cell signaling pathways. Here, we focused on the desmosomal adhesion molecules Desmoglein 2 (Dsg2) and Desmocollin 2 (Dsc2), and their contribution to migration and invasion in pancreatic cancer cells. Silencing of Dsg2 but not Dsc2 resulted in loss of cell cohesion and enhanced migration, and invasion of pancreatic adenocarcinoma cells. To identify potential pathways regulated by Dsg2, we performed kinase arrays and detected the activity of ERK and growth factor receptors to be significantly enhanced in Dsg2-deficient cells. Consequently, inhibition of ERK phosphorylation in Dsg2 knockdown cells normalized migration. Loss of Dsg2 resulted in reduced levels of the desmosomal adapter protein and transcriptional regulator Plakoglobin (PG) in an ERK-dependent manner, whereas other desmosomal molecules were not altered. Overexpression of PG rescued enhanced migration induced by silencing of Dsg2. These results identify a novel pro-migratory pathway of pancreatic cancer cells in which loss of Dsg2 reduces the levels of PG via deregulated MAPK signaling.
Collapse
Affiliation(s)
- Katharina Hütz
- Department I, Institute of Anatomy and Cell Biology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Julian Zeiler
- Department I, Institute of Anatomy and Cell Biology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Lena Sachs
- Department I, Institute of Anatomy and Cell Biology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Steffen Ormanns
- Institute of Pathology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Volker Spindler
- Department I, Institute of Anatomy and Cell Biology, Ludwig-Maximilians-Universität Munich, Munich, Germany
| |
Collapse
|
48
|
Schinner C, Vielmuth F, Rötzer V, Hiermaier M, Radeva MY, Co TK, Hartlieb E, Schmidt A, Imhof A, Messoudi A, Horn A, Schlipp A, Spindler V, Waschke J. Adrenergic Signaling Strengthens Cardiac Myocyte Cohesion. Circ Res 2017; 120:1305-1317. [PMID: 28289018 DOI: 10.1161/circresaha.116.309631] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 02/22/2017] [Accepted: 03/10/2017] [Indexed: 01/08/2023]
Abstract
RATIONALE The sympathetic nervous system is a major mediator of heart function. Intercalated discs composed of desmosomes, adherens junctions, and gap junctions provide the structural backbone for coordinated contraction of cardiac myocytes. OBJECTIVE Gap junctions dynamically remodel to adapt to sympathetic signaling. However, it is unknown whether such rapid adaption also occurs for the adhesive function provided by desmosomes and adherens junctions. METHODS AND RESULTS Atomic force microscopy revealed that β-adrenergic signaling enhances both the number of desmoglein 2-specific interactions along cell junctions and the mean desmoglein 2-mediated binding forces, whereas N-cadherin-mediated interactions were not affected. This was accompanied by increased cell cohesion in cardiac myocyte cultures and murine heart slices. Enhanced desmoglein 2-positive contacts and increased junction length as revealed by immunofluorescence and electron microscopy reflected cAMP-induced reorganization of intercellular contacts. The mechanism underlying cAMP-mediated strengthening of desmoglein 2 binding was dependent on expression of the intercalated disc plaque protein plakoglobin (Pg) and direct phosphorylation at S665 by protein kinase A: Pg deficiency as well as overexpression of the phospho-deficient Pg-mutant S665A abrogated both cAMP-mediated junctional remodeling and increase of cohesion. Moreover, Pg knockout hearts failed to functionally adapt to adrenergic stimulation. CONCLUSIONS Taken together, we provide first evidence for positive adhesiotropy as a new cardiac function of sympathetic signaling. Positive adhesiotropy is dependent on Pg phosphorylation at S665 by protein kinase A. This mechanism may be of high medical relevance because loss of junctional Pg is a hallmark of arrhythmogenic cardiomyopathy.
Collapse
Affiliation(s)
- Camilla Schinner
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Franziska Vielmuth
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Vera Rötzer
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Matthias Hiermaier
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Mariya Y Radeva
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Thu Kim Co
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Eva Hartlieb
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Andreas Schmidt
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Axel Imhof
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Ahmed Messoudi
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Anja Horn
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Angela Schlipp
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Volker Spindler
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany
| | - Jens Waschke
- From the Institute of Anatomy and Cell Biology (C.S., F.V., V.R., M.H., M.Y.R., T.K.C., E.H., A.M., A.H., A. Schlipp, V.S., J.W.) and Biomedical Center and Center for Integrated Protein Sciences Munich (A. Schmidt, A.I.), Ludwig-Maximilians-Universität, Germany.
| |
Collapse
|
49
|
van Opbergen CJM, Delmar M, van Veen TAB. Potential new mechanisms of pro-arrhythmia in arrhythmogenic cardiomyopathy: focus on calcium sensitive pathways. Neth Heart J 2017; 25:157-169. [PMID: 28102477 PMCID: PMC5313453 DOI: 10.1007/s12471-017-0946-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Arrhythmogenic cardiomyopathy, or its most well-known subform arrhythmogenic right ventricular cardiomyopathy (ARVC), is a cardiac disease mainly characterised by a gradual replacement of the myocardial mass by fibrous and fatty tissue, leading to dilatation of the ventricular wall, arrhythmias and progression towards heart failure. ARVC is commonly regarded as a disease of the intercalated disk in which mutations in desmosomal proteins are an important causative factor. Interestingly, the Dutch founder mutation PLN R14Del has been identified to play an additional, and major, role in ARVC patients within the Netherlands. This is remarkable since the phospholamban (PLN) protein plays a leading role in regulation of the sarcoplasmic reticulum calcium load rather than in the establishment of intercellular integrity. In this review we outline the intracellular cardiac calcium dynamics and relate pathophysiological signalling, induced by disturbed calcium handling, with activation of calmodulin dependent kinase II (CaMKII) and calcineurin A (CnA). We postulate a thus far unrecognised role for Ca2+ sensitive signalling proteins in maladaptive remodelling of the macromolecular protein complex that forms the intercalated disk, during pro-arrhythmic remodelling of the heart.
Collapse
Affiliation(s)
- C J M van Opbergen
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M Delmar
- The Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, USA
| | - T A B van Veen
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
50
|
Moftah H, Dias K, Apu EH, Liu L, Uttagomol J, Bergmeier L, Kermorgant S, Wan H. Desmoglein 3 regulates membrane trafficking of cadherins, an implication in cell-cell adhesion. Cell Adh Migr 2016; 11:211-232. [PMID: 27254775 DOI: 10.1080/19336918.2016.1195942] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
E-cadherin mediated cell-cell adhesion plays a critical role in epithelial cell polarization and morphogenesis. Our recent studies suggest that the desmosomal cadherin, desmoglein 3 (Dsg3) cross talks with E-cadherin and regulates its adhesive function in differentiating keratinocytes. However, the underlying mechanism remains not fully elucidated. Since E-cadherin trafficking has been recognized to be a central determinant in cell-cell adhesion and homeostasis we hypothesize that Dsg3 may play a role in regulating E-cadherin trafficking and hence the cell-cell adhesion. Here we investigated this hypothesis in cells with loss of Dsg3 function through RNAi mediated Dsg3 knockdown or the stable expression of the truncated mutant Dsg3ΔC. Our results showed that loss of Dsg3 resulted in compromised cell-cell adhesion and reduction of adherens junction and desmosome protein expression as well as the cortical F-actin formation. As a consequence, cells failed to polarize but instead displayed aberrant cell flattening. Furthermore, retardation of E-cadherin internalization and recycling was consistently observed in these cells during the process of calcium induced junction assembling. In contrast, enhanced cadherin endocytosis was detected in cells with overexpression of Dsg3 compared to control cells. Importantly, this altered cadherin trafficking was found to be coincided with the reduced expression and activity of Rab proteins, including Rab5, Rab7 and Rab11 which are known to be involved in E-cadherin trafficking. Taken together, our findings suggest that Dsg3 functions as a key in cell-cell adhesion through at least a mechanism of regulating E-cadherin membrane trafficking.
Collapse
Affiliation(s)
- Hanan Moftah
- a Centre for Clinical and Diagnostic Oral Sciences, Institute of Dentistry, Barts and The London, School of Medicine and Dentistry , Queen Mary University of London , Whitechapel, London , UK
| | - Kasuni Dias
- a Centre for Clinical and Diagnostic Oral Sciences, Institute of Dentistry, Barts and The London, School of Medicine and Dentistry , Queen Mary University of London , Whitechapel, London , UK
| | - Ehsanul Hoque Apu
- a Centre for Clinical and Diagnostic Oral Sciences, Institute of Dentistry, Barts and The London, School of Medicine and Dentistry , Queen Mary University of London , Whitechapel, London , UK
| | - Li Liu
- a Centre for Clinical and Diagnostic Oral Sciences, Institute of Dentistry, Barts and The London, School of Medicine and Dentistry , Queen Mary University of London , Whitechapel, London , UK
| | - Jutamas Uttagomol
- a Centre for Clinical and Diagnostic Oral Sciences, Institute of Dentistry, Barts and The London, School of Medicine and Dentistry , Queen Mary University of London , Whitechapel, London , UK
| | - Lesley Bergmeier
- a Centre for Clinical and Diagnostic Oral Sciences, Institute of Dentistry, Barts and The London, School of Medicine and Dentistry , Queen Mary University of London , Whitechapel, London , UK
| | - Stephanie Kermorgant
- b Barts Cancer Institute, John Vane Science Center , Charterhouse Square, London , UK
| | - Hong Wan
- a Centre for Clinical and Diagnostic Oral Sciences, Institute of Dentistry, Barts and The London, School of Medicine and Dentistry , Queen Mary University of London , Whitechapel, London , UK
| |
Collapse
|