1
|
Chen X, Xu MN, Wu ZQ, Huang R, Lu HY, Peng X, Zeng K, Li CX. JAK1/2 Inhibitors Alleviate the Damage of Intercellular Adhesion by Reducing Endoplasmic Reticulum Stress-Induced Apoptosis in Pemphigus Vulgaris. Exp Dermatol 2025; 34:e70121. [PMID: 40392073 DOI: 10.1111/exd.70121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 04/25/2025] [Accepted: 04/27/2025] [Indexed: 05/22/2025]
Abstract
Pemphigus vulgaris (PV), a severe autoimmune disease with high morbidity and mortality, necessitates innovative therapies to improve outcomes while minimising the adverse effects of conventional immunosuppressants. Immunohistochemical analysis revealed elevated phosphorylated Janus kinase (p-JAK)1 and p-JAK2 expression in PV lesions, complemented by transcriptome data showing JAK/STAT pathway dysregulation. Using a PV acantholysis model, we demonstrated that Ruxolitinib, a JAK1/2 inhibitor, significantly reduced keratinocyte apoptosis, enhanced cell adhesion, and alleviated endoplasmic reticulum (ER) stress. Additionally, Ruxolitinib mitigated tunicamycin-induced ER stress and apoptosis in HaCaT cells. These findings establish a crucial role for JAK1/2 in PV pathogenesis, demonstrating that their inhibition alleviates ER stress, reduces apoptosis, and improves cell adhesion. Our results provide a theoretical foundation for the clinical application of JAK inhibitors in PV treatment.
Collapse
Affiliation(s)
- Xi Chen
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mei-Nian Xu
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zheng-Quan Wu
- Department of Dermatology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Rong Huang
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hong-Yan Lu
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoming Peng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kang Zeng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chang-Xing Li
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Dermatology, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, China
| |
Collapse
|
2
|
Peng X, Wang S, Wu K, Cook C, Li L, Wang Z, Gu H, Lu M, Hu G, Ren K, Hu G, Zeng W, Xia Y, Liu Y. Effect of opioid receptor antagonist on mitigating tumor necrosis factor-like weak inducer of apoptosis (TWEAK)-induced apoptolysis in pemphigus pathogenesis. J Autoimmun 2024; 149:103307. [PMID: 39276627 DOI: 10.1016/j.jaut.2024.103307] [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: 04/23/2024] [Revised: 08/13/2024] [Accepted: 08/29/2024] [Indexed: 09/17/2024]
Abstract
Pemphigus is a severe autoimmune blistering disease characterized by acantholysis triggered by autoantibodies against desmoglein 1 and 3 (DSG1/3). Apoptosis plays a pivotal role in facilitating acantholysis, yet the precise underlying mechanism remains obscure. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is known to promote apoptosis and disrupt cell junctions, although its involvement in pemphigus pathogenesis remains ambiguous. Our study observed decreased DSG1/3 expression alongside increased TWEAK/fibroblast growth factor-inducible 14 (Fn14) expression and keratinocyte apoptosis in both lesional and perilesional skin. In vitro experiments revealed that TWEAK-stimulated keratinocytes exhibited enhanced apoptosis, STAT1 phosphorylation, and reduced intercellular DSG1/3 expression. Notably, bulk-RNA sequencing unveiled that CASPASE-3 was responsible for mediating the DSG1/3 depletion, as confirmed by direct interaction with DSG1/3 in a co-immunoprecipitation assay. Naloxone, known for preserving cellular adhesion and preventing cell death, effectively reduced apoptosis and restored DSG1/3 levels in TWEAK-stimulated keratinocytes. The anti-apoptotic properties of naloxone were further validated in a murine pemphigus model. Our findings elucidate that TWEAK facilitates keratinocyte apoptosis by augmenting caspase-3 activity, leading to DSG1/3 depletion and apoptosis in pemphigus. Importantly, naloxone can counter TWEAK-induced apoptosis in pemphigus pathogenesis, offering a potential therapeutic intervention.
Collapse
Affiliation(s)
- Xueting Peng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Sijia Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Kunyi Wu
- Core Research Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Christopher Cook
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Liang Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Zhao Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Hanjiang Gu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Mei Lu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Guanglei Hu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Kaixuan Ren
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Gang Hu
- Department of Dermatology, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Weihui Zeng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China.
| | - Yale Liu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China.
| |
Collapse
|
3
|
Mueller EJ, Rahimi S, Sauta P, Shojaeian T, Durrer L, Quinche S, Francois M, Locher E, Edler M, Illi M, Gentinetta T, Lau K, Pojer F, Borradori L, Hariton WVJ. Standardized Production of Anti-Desmoglein 3 Antibody AK23 for Translational Pemphigus Vulgaris Research. Curr Protoc 2024; 4:e1118. [PMID: 39169810 DOI: 10.1002/cpz1.1118] [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] [Indexed: 08/23/2024]
Abstract
Antibody-mediated receptor activation is successfully used to develop medical treatments. If the activation induces a pathological response, such antibodies are also excellent tools for defining molecular mechanisms of target receptor malfunction and designing rescue therapies. Prominent examples are naturally occurring autoantibodies inducing the severe blistering disease pemphigus vulgaris (PV). In the great majority of patients, the antibodies bind to the adhesion receptor desmoglein 3 (Dsg3) and interfere with cell signaling to provoke severe blistering in the mucous membranes and/or skin. The identification of a comprehensive causative signaling network downstream of antibody-targeted Dsg3 receptors (e.g., shown by pharmacological activators or inhibitors) is currently being discussed as a basis to develop urgently needed first-line treatments for PV patients. Although polyclonal PV IgG antibodies have been used as proof of principle for pathological signal activation, monospecific anti-Dsg3 antibodies are necessary and have been developed to identify pathological Dsg3 receptor-mediated signal transduction. The experimental monospecific PV antibody AK23, produced from hybridoma cells, was extensively tested in our laboratory in both in vitro and in vivo models for PV and proved to recapitulate the clinicopathological features of PV when generated using the standardized production and purification protocols described herein. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Bovine IgG stripping from FBS and quality control Basic Protocol 2: AK23 hybridoma expansion and IgG production Basic Protocol 3: AK23 IgG purification Basic Protocol 4: AK23 IgG quality control Support Protocol 1: Detection of endotoxin levels Support Protocol 2: Detection and removal of mycoplasma.
Collapse
Affiliation(s)
- Eliane J Mueller
- Department for Biomedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Bern, Switzerland
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Siavash Rahimi
- Department for Biomedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Bern, Switzerland
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Patrizia Sauta
- Department for Biomedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Bern, Switzerland
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Taravat Shojaeian
- Department for Biomedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Bern, Switzerland
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Laurence Durrer
- Protein Production and Structure Core Facility, School of Life Sciences, EPFL Lausanne, Lausanne, Switzerland
| | - Soraya Quinche
- Protein Production and Structure Core Facility, School of Life Sciences, EPFL Lausanne, Lausanne, Switzerland
| | - Michael Francois
- Protein Production and Structure Core Facility, School of Life Sciences, EPFL Lausanne, Lausanne, Switzerland
| | - Elisabeth Locher
- CSL, CSL Biologics Research Centre, Bern, Switzerland
- Swiss Institute for Translational and Entrepreneurial Medicine, sitem-insel, Bern, Switzerland
| | - Monika Edler
- CSL, CSL Biologics Research Centre, Bern, Switzerland
- Swiss Institute for Translational and Entrepreneurial Medicine, sitem-insel, Bern, Switzerland
| | - Marlies Illi
- CSL, CSL Biologics Research Centre, Bern, Switzerland
- Swiss Institute for Translational and Entrepreneurial Medicine, sitem-insel, Bern, Switzerland
| | - Thomas Gentinetta
- CSL, CSL Biologics Research Centre, Bern, Switzerland
- Swiss Institute for Translational and Entrepreneurial Medicine, sitem-insel, Bern, Switzerland
| | - Kelvin Lau
- Protein Production and Structure Core Facility, School of Life Sciences, EPFL Lausanne, Lausanne, Switzerland
| | - Florence Pojer
- Protein Production and Structure Core Facility, School of Life Sciences, EPFL Lausanne, Lausanne, Switzerland
| | - Luca Borradori
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - William V J Hariton
- Department for Biomedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Bern, Switzerland
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
4
|
Hariton WV, Schulze K, Rahimi S, Shojaeian T, Feldmeyer L, Schwob R, Overmiller AM, Sayar BS, Borradori L, Mahoney MG, Galichet A, Müller EJ. A desmosomal cadherin controls multipotent hair follicle stem cell quiescence and orchestrates regeneration through adhesion signaling. iScience 2023; 26:108568. [PMID: 38162019 PMCID: PMC10755723 DOI: 10.1016/j.isci.2023.108568] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/03/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2024] Open
Abstract
Stem cells (SCs) are critical to maintain tissue homeostasis. However, it is currently not known whether signaling through cell junctions protects quiescent epithelial SC reservoirs from depletion during disease-inflicted damage. Using the autoimmune model disease pemphigus vulgaris (PV), this study reveals an unprecedented role for a desmosomal cadherin in governing SC quiescence and regeneration through adhesion signaling in the multipotent mouse hair follicle compartment known as the bulge. Autoantibody-mediated, mechanical uncoupling of desmoglein (Dsg) 3 transadhesion activates quiescent bulge SC which lose their multipotency and stemness, become actively cycling, and finally delaminate from their epithelial niche. This then initiates a self-organized regenerative program which restores Dsg3 function and bulge morphology including SC quiescence and multipotency. These profound changes are triggered by the sole loss of functional Dsg3, resemble major signaling events in Dsg3-/- mice, and are driven by SC-relevant EGFR activation and Wnt modulation requiring longitudinal repression of Hedgehog signaling.
Collapse
Affiliation(s)
- William V.J. Hariton
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Katja Schulze
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Siavash Rahimi
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
| | - Taravat Shojaeian
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
| | - Laurence Feldmeyer
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Roman Schwob
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Andrew M. Overmiller
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Beyza S. Sayar
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
| | - Luca Borradori
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
| | - Mỹ G. Mahoney
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Arnaud Galichet
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Eliane J. Müller
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, 3008 Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, 3008 Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
5
|
Hartmann V, Hariton WV, Rahimi S, Hammers CM, Ludwig RJ, Müller EJ, Hundt JE. The human skin organ culture model as an optimal complementary tool for murine pemphigus models. Lab Anim 2023; 57:381-395. [PMID: 36647613 DOI: 10.1177/00236772221145647] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Pemphigus is a severe autoimmune bullous disease of the skin and/or mucous membranes caused by autoantibodies that mainly target the adhesion proteins desmoglein (Dsg) 3 and/or Dsg1. Clinically, pemphigus is characterized by flaccid blistering, leading to severe water and electrolyte loss. Before the introduction of corticosteroid treatment, the disease turned out to be fatal in many cases. Despite recent therapeutic improvements, treatment of pemphigus patients is centred on prolonged systemic immunosuppression and remains challenging. Current drug development for pemphigus has a strong focus on disease-causing B cells and autoantibodies and, more recently, also on modulating autoantibody-induced tissue pathology and keratinocyte signalling. This drug development requires reliable pre-clinical model systems replicating the pathogenesis of the human disease. Among those are neonatal and adult mouse models based on the transfer of Dsg3, Dsg1/3 or Dsg1-specific autoantibodies. To reduce the number of animal experiments, we recently established a standardized human skin organ culture (HSOC) model for pemphigus. This model reproduces the clinical phenotype of autoantibody-induced tissue pathology in pemphigus vulgaris. For induction of blistering, a recombinant single-chain variable fragment (scFv) targeting both Dsg1 and 3 is injected into pieces of human skin (obtained from plastic surgeries). Further characterization of the HSOC model demonstrated that key morphologic, molecular and immunologic features of pemphigus are being replicated. Thus, the pemphigus HSOC model is an excellent alternative to pemphigus animal model systems that are based on the transfer of (auto)antibodies.
Collapse
Affiliation(s)
- Veronika Hartmann
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Germany
| | - William Vj Hariton
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Switzerland
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Switzerland
| | - Siavash Rahimi
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Switzerland
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Switzerland
| | | | - Ralf J Ludwig
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Germany
- Centre for Research on Inflammation of the Skin, University of Lübeck, Germany
- Department of Dermatology, Allergy, and Venerology, University of Lübeck, Germany
| | - Eliane J Müller
- Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Switzerland
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Switzerland
| | - Jennifer E Hundt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Germany
- Centre for Research on Inflammation of the Skin, University of Lübeck, Germany
| |
Collapse
|
6
|
Duan J, Grando C, Liu S, Chernyavsky A, Chen JK, Andersen B, Grando SA. The M3 Muscarinic Acetylcholine Receptor Promotes Epidermal Differentiation. J Invest Dermatol 2022; 142:3211-3221.e2. [PMID: 35870560 PMCID: PMC9851810 DOI: 10.1016/j.jid.2022.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 01/21/2023]
Abstract
The M3 muscarinic acetylcholine receptor is predominantly expressed in the basal epidermal layer where it mediates the effects of the autocrine/paracrine cytotransmitter acetylcholine. Patients with the autoimmune blistering disease pemphigus develop autoantibodies to M3 muscarinic acetylcholine receptor and show alterations in keratinocyte adhesion, proliferation, and differentiation, suggesting that M3 muscarinic acetylcholine receptor controls these cellular functions. Chmr3-/- mice display altered epidermal morphology resembling that seen in patients with pemphigus vulgaris. In this study, we characterized the cellular and molecular mechanisms through which M3 muscarinic acetylcholine receptor controls epidermal structure and function. We used single-cell RNA sequencing to evaluate keratinocyte heterogeneity and identify differentially expressed genes in specific subpopulations of epidermal cells in Chmr3-/- neonatal mice. We found that Chmr3-/- mice feature abnormal epidermal morphology characterized by accumulation of nucleated basal cells, shrinkage of basal keratinocytes, and enlargement of intercellular spaces. These morphologic changes were associated with upregulation of cell proliferation genes and downregulation of genes contributing to epidermal differentiation, extracellular matrix formation, intercellular adhesion, and cell arrangement. These findings provide, to our knowledge, previously unreported insights into how acetylcholine controls epidermal differentiation and lay a groundwork for future translational studies evaluating the therapeutic potential of cholinergic drugs in dermatology.
Collapse
Affiliation(s)
- Junyan Duan
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA,NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California, USA
| | - Charles Grando
- Department of Dermatology, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Shuman Liu
- Division of Endocrinology, Department of Medicine, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Alex Chernyavsky
- Department of Dermatology, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Jefferson K. Chen
- Division of Endocrinology, Department of Medicine, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Bogi Andersen
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA,NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California, USA,Division of Endocrinology, Department of Medicine, School of Medicine, University of California, Irvine, Irvine, California, USA,Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Sergei A. Grando
- Department of Dermatology, School of Medicine, University of California, Irvine, Irvine, California, USA,Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, California, USA,Institute for Immunology, University of California, Irvine, Irvine, California, USA
| |
Collapse
|
7
|
Apoptolysis: a less understood concept in the pathogenesis of Pemphigus Vulgaris. Apoptosis 2022; 27:322-328. [PMID: 35445279 DOI: 10.1007/s10495-022-01726-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2022] [Indexed: 11/02/2022]
Abstract
Pemphigus Vulgaris (PV) is a severe autoimmune disease characterized by supra-basal blisters in the skin and mucous membranes of a wide range of mammals, including humans. It not only affects the skin but also has severe oral manifestations. It has been stated that auto-antibodies are produced, for unknown reasons, which are directed against desmogleins present on the epithelium and thus leads to acantholysis and intraepithelial blistering. But the exact mechanism is still not completely understood. Here we would like to shed light on a new pathologic mechanism i.e., apoptolysis, which emphasizes that apoptotic enzymes contribute to acantholysis development both in terms of molecular events and chronologic sequence. A possible role of apoptolysis has been discussed in purview of PV.
Collapse
|
8
|
Chernyavsky A, Khylynskyi MM, Patel KG, Grando SA. Chronic exposure to the anti-M3 muscarinic acetylcholine receptor autoantibody in pemphigus vulgaris contributes to disease pathophysiology. J Biol Chem 2022; 298:101687. [PMID: 35143842 PMCID: PMC8897697 DOI: 10.1016/j.jbc.2022.101687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 12/31/2022] Open
Abstract
Pemphigus vulgaris (PV) is a potentially lethal autoimmune mucocutaneous blistering disease characterized by binding of IgG autoantibodies (AuAbs) to keratinocytes (KCs). In addition to AuAbs against adhesion molecules desmogleins 1 and 3, PV patients also produce an AuAb against the M3 muscarinic acetylcholine (ACh) receptor (M3AR) that plays an important role in regulation of vital functions of KCs upon binding endogenous ACh. This anti-M3AR AuAb is pathogenic because its adsorption eliminates the acantholytic activity of PV IgG; however, the molecular mechanism of its action is unclear. In the present study, we sought to elucidate the mode of immunopharmacologic action of the anti-M3AR AuAb in PV. Short-term exposures of cultured KCs to PV IgG or the muscarinic agonist muscarine both induced changes in the expression of keratins 5 and 10, consistent with the inhibition of proliferation and upregulated differentiation and in keeping with the biological function of M3AR. In contrast, long-term incubations induced a keratin expression pattern consistent with upregulated proliferation and decreased differentiation, in keeping with the hyperproliferative state of KCs in PV. This change could result from desensitization of the M3AR, representing the net antagonist-like effect of the AuAb. Therefore, chronic exposure of KCs to the anti-M3AR AuAb interrupts the physiological regulation of KCs by endogenous ACh, contributing to the onset of acantholysis. Since cholinergic agents have already demonstrated antiacantholytic activity in a mouse model of PV and in PV patients, our results have translational significance and can guide future development of therapies for PV patients employing cholinergic drugs.
Collapse
Affiliation(s)
- Alex Chernyavsky
- Department of Dermatology, University of California Irvine, Irvine, California, USA
| | | | - Krupa G Patel
- Department of Neurology, Oakland University William Beaumont School of Medicine, Rochester, Michigan, USA
| | - Sergei A Grando
- Department of Dermatology, University of California Irvine, Irvine, California, USA; Department of Biological Chemistry, University of California Irvine, Irvine, California, USA; Institute for Immunology, University of California Irvine, Irvine, California, USA.
| |
Collapse
|
9
|
Bumiller-Bini Hoch V, Schneider L, Pumpe AE, Lüders E, Hundt JE, Boldt ABW. Marked to Die-Cell Death Mechanisms for Keratinocyte Acantholysis in Pemphigus Diseases. Life (Basel) 2022; 12:life12030329. [PMID: 35330080 PMCID: PMC8948972 DOI: 10.3390/life12030329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 11/17/2022] Open
Abstract
Pemphigus is a group of blistering autoimmune diseases causing painful skin lesions, characterized by acantholysis and by the production of autoantibodies against, mainly, adhesion proteins. We reviewed the literature for molecules and/ or features involved in the 12 cell death pathways described by Nomenclature Committee on Cell Death, taking place in pemphigus patients, cell lines, or human skin organ cultures treated with sera or IgG from pemphigus patients or in pemphigus mouse models, and found 61 studies mentioning 97 molecules involved in cell death pathways. Among the molecules, most investigated were pleiotropic molecules such as TNF and CASP3, followed by FASL and CASP8, and then by FAS, BAX, BCL2, and TP53, all involved in more than one pathway but interpreted to function only within apoptosis. Most of these previous investigations focused only on apoptosis, but four recent studies, using TUNEL assays and/or electron microscopy, disqualified this pathway as a previous event of acantholysis. For PV, apoptolysis was suggested as a cell death mechanism based on pathogenic autoantibodies diversity, mitochondrial dysfunction, and p38 MAPK signaling. To answer those many questions that remain on cell death and pemphigus, we propose well-controlled, statistically relevant investigations on pemphigus and cell death pathways besides apoptosis, to overcome the challenges of understanding the etiopathology of pemphigus diseases.
Collapse
Affiliation(s)
- Valéria Bumiller-Bini Hoch
- Laboratory of Human Molecular Genetics, Department of Genetics, Federal University of Paraná, Curitiba 81531-980, Brazil; (V.B.-B.H.); (L.S.)
- Postgraduate Program in Genetics, Department of Genetics, Federal University of Paraná (UFPR), Curitiba 81531-980, Brazil
- Lübeck Institute of Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany; (A.E.P.); (E.L.); (J.E.H.)
| | - Larissa Schneider
- Laboratory of Human Molecular Genetics, Department of Genetics, Federal University of Paraná, Curitiba 81531-980, Brazil; (V.B.-B.H.); (L.S.)
| | - Anna Elisabeth Pumpe
- Lübeck Institute of Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany; (A.E.P.); (E.L.); (J.E.H.)
| | - Emelie Lüders
- Lübeck Institute of Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany; (A.E.P.); (E.L.); (J.E.H.)
| | - Jennifer Elisabeth Hundt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany; (A.E.P.); (E.L.); (J.E.H.)
| | - Angelica Beate Winter Boldt
- Laboratory of Human Molecular Genetics, Department of Genetics, Federal University of Paraná, Curitiba 81531-980, Brazil; (V.B.-B.H.); (L.S.)
- Correspondence:
| |
Collapse
|
10
|
Huda S, Chau B, Chen C, Somal H, Chowdhury N, Cirillo N. Caspase Inhibition as a Possible Therapeutic Strategy for Pemphigus Vulgaris: A Systematic Review of Current Evidence. BIOLOGY 2022; 11:biology11020314. [PMID: 35205180 PMCID: PMC8869094 DOI: 10.3390/biology11020314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/03/2022] [Accepted: 02/10/2022] [Indexed: 12/09/2022]
Abstract
Simple Summary Pemphigus vulgaris is a potentially fatal disease characterised by blister formation affecting the skin and mouth. The mechanisms of blister formation may involve a biological process called apoptosis—a type of cell death—and some death-associated molecules known as caspases. Our review of the existing literature shows that caspase inhibitors exhibit an inhibitory effect on PV-induced apoptosis formation in vitro. In particular, activity of caspase 1 and caspase 3 is essential for the development of PV in vitro and in vivo. However, a majority of in vivo studies assessing caspase inhibition in PV models have a high risk of bias. Abstract Background: Pemphigus vulgaris (PV) is an IgG-mediated autoimmune disease characterised by epithelial cell–cell detachment (acantholysis) resulting in mucocutaneous blistering. The exact pathogenesis of blister formation is unknown and this has hampered the development of non-steroidal, mechanism-based treatments for this autoimmune disease. This systematic review aims to investigate the role of caspases in the pathogenesis of PV to inform the choice of more targeted therapeutic agents. Methods: A systematic search of MEDLINE/PubMed and Scopus databases was conducted to identify eligible studies. Multiple phases of inclusion and exclusion of the primary articles were conducted in pairs, and studies were recorded and analysed according to the latest version of the preferred reporting items for systematic reviews and meta-analyses (PRISMA). Risk of bias assessment was conducted for extracted in vivo animal intervention studies using SYRCLE’s risk of bias tool. Results: Eight articles from a total of 2338 in vitro, in vivo, and human studies met the inclusion criteria, with a high degree of inter-rater reliability. By and large, the results show that caspase activation was pathogenic in experimental PV because pan-caspase inhibitors could block or reduce PV acantholysis and blistering in vitro and in vivo, respectively. The pathogenic pathways identified involved caspase-1 and caspase-3. One study failed to show any improvement in the PV model with a caspase inhibitor. The majority of animal studies had high or unclear risk of bias. Conclusion: There are consistent data pointing towards a pathogenic role of caspase activation in PV acantholysis. However, high-quality evidence to confirm that caspase inhibition can prevent PV-induced blistering in vivo is limited. Therefore, further research is required to test the preclinical efficacy of caspase inhibitors in PV.
Collapse
|
11
|
Yin L, Li Q, Mrdenovic S, Chu GCY, Wu BJ, Bu H, Duan P, Kim J, You S, Lewis MS, Liang G, Wang R, Zhau HE, Chung LWK. KRT13 promotes stemness and drives metastasis in breast cancer through a plakoglobin/c-Myc signaling pathway. Breast Cancer Res 2022; 24:7. [PMID: 35078507 PMCID: PMC8788068 DOI: 10.1186/s13058-022-01502-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 01/13/2022] [Indexed: 02/08/2023] Open
Abstract
Background Keratins (KRTs) are intermediate filament proteins that interact with multiple regulatory proteins to initiate signaling cascades. Keratin 13 (KRT13) plays an important role in breast cancer progression and metastasis. The objective of this study is to elucidate the mechanism by which KRT13 promotes breast cancer growth and metastasis.
Methods The function and mechanisms of KRT13 in breast cancer progression and metastasis were assessed by overexpression and knockdown followed by examination of altered behaviors in breast cancer cells and in xenograft tumor formation in mouse mammary fat pad. Human breast cancer specimens were examined by immunohistochemistry and multiplexed quantum dot labeling analysis to correlate KRT13 expression to breast cancer progression and metastasis. Results KRT13-overexpressing MCF7 cells displayed increased proliferation, invasion, migration and in vivo tumor growth and metastasis to bone and lung. Conversely, KRT13 knockdown inhibited the aggressive behaviors of HCC1954 cells. At the molecular level, KRT13 directly interacted with plakoglobin (PG, γ-catenin) to form complexes with desmoplakin (DSP). This complex interfered with PG expression and nuclear translocation and abrogated PG-mediated suppression of c-Myc expression, while the KRT13/PG/c-Myc signaling pathway increased epithelial to mesenchymal transition and stem cell-like phenotype. KRT13 expression in 58 human breast cancer tissues was up-regulated especially at the invasive front and in metastatic specimens (12/18) (p < 0.05). KRT13 up-regulation in primary breast cancer was associated with decreased overall patient survival. Conclusions This study reveals that KRT13 promotes breast cancer cell growth and metastasis via a plakoglobin/c-Myc pathway. Our findings reveal a potential novel pathway for therapeutic targeting of breast cancer progression and metastasis. Supplementary Information The online version contains supplementary material available at 10.1186/s13058-022-01502-6.
Collapse
Affiliation(s)
- Lijuan Yin
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA
| | - Qinlong Li
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA
| | - Stefan Mrdenovic
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA
| | - Gina Chia-Yi Chu
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA
| | - Boyang Jason Wu
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA
| | - Hong Bu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Peng Duan
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA
| | - Jayoung Kim
- Division of Cancer Biology and Therapeutics, Departments of Surgery and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sungyong You
- Division of Cancer Biology and Therapeutics, Departments of Surgery and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael S Lewis
- Department of Pathology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Gangning Liang
- Department of Urology, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Ruoxiang Wang
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA.
| | - Haiyen E Zhau
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA
| | - Leland W K Chung
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, 8750 Beverly Boulevard, Atrium 105, Los Angeles, CA, 90048, USA
| |
Collapse
|
12
|
Müller L, Hatzfeld M, Keil R. Desmosomes as Signaling Hubs in the Regulation of Cell Behavior. Front Cell Dev Biol 2021; 9:745670. [PMID: 34631720 PMCID: PMC8495202 DOI: 10.3389/fcell.2021.745670] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 08/31/2021] [Indexed: 12/19/2022] Open
Abstract
Desmosomes are intercellular junctions, which preserve tissue integrity during homeostatic and stress conditions. These functions rely on their unique structural properties, which enable them to respond to context-dependent signals and transmit them to change cell behavior. Desmosome composition and size vary depending on tissue specific expression and differentiation state. Their constituent proteins are highly regulated by posttranslational modifications that control their function in the desmosome itself and in addition regulate a multitude of desmosome-independent functions. This review will summarize our current knowledge how signaling pathways that control epithelial shape, polarity and function regulate desmosomes and how desmosomal proteins transduce these signals to modulate cell behavior.
Collapse
Affiliation(s)
- Lisa Müller
- Department for Pathobiochemistry, Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Mechthild Hatzfeld
- Department for Pathobiochemistry, Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - René Keil
- Department for Pathobiochemistry, Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Huang Y, Jedličková H, Cai Y, Rehman A, Gammon L, Ahmad US, Uttagomol J, Parkinson EK, Fortune F, Wan H. Oxidative Stress-Mediated YAP Dysregulation Contributes to the Pathogenesis of Pemphigus Vulgaris. Front Immunol 2021; 12:649502. [PMID: 33968042 PMCID: PMC8098436 DOI: 10.3389/fimmu.2021.649502] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/29/2021] [Indexed: 11/15/2022] Open
Abstract
Pemphigus Vulgaris (PV) is a life-threatening autoimmune disease manifested with blisters in the skin and mucosa and caused by autoantibodies against adhesion protein desmoglein-3 (Dsg3) expressed in epithelial membrane linings of these tissues. Despite many studies, the pathogenesis of PV remains incompletely understood. Recently we have shown Dsg3 plays a role in regulating the yes-associated protein (YAP), a co-transcription factor and mechanical sensor, and constraining reactive oxygen species (ROS). This study investigated the effect of PV sera as well as the anti-Dsg3 antibody AK23 on these molecules. We detected elevated YAP steady-state protein levels in PV cells surrounding blisters and perilesional regions and in keratinocytes treated with PV sera and AK23 with concomitant transient ROS overproduction. Cells treated with hydrogen peroxide also exhibited augmented nuclear YAP accompanied by reduction of Dsg3 and α-catenin, a negative regulator of YAP. As expected, transfection of α-catenin-GFP plasmid rendered YAP export from the nucleus evoked by hydrogen peroxide. In addition, suppression of total YAP was observed in hydrogen peroxide treated cells exposed to antioxidants with enhanced cell-cell adhesion being confirmed by decreased fragmentation in the dispase assay compared to hydrogen peroxide treatment alone. On the other hand, the expression of exogenous YAP disrupted intercellular junction assembly. In contrast, YAP depletion resulted in an inverse effect with augmented expression of junction assembly proteins, including Dsg3 and α-catenin capable of abolishing the effect of AK23 on Dsg3 expression. Finally, inhibition of other kinase pathways, including p38MAPK, also demonstrated suppression of YAP induced by hydrogen peroxide. Furthermore, antioxidant treatment of keratinocytes suppressed PV sera-induced total YAP accumulation. In conclusion, this study suggests that oxidative stress coupled with YAP dysregulation attributes to PV blistering, implying antioxidants may be beneficial in the treatment of PV.
Collapse
Affiliation(s)
- Yunying Huang
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Hana Jedličková
- Department of Dermatology, St. Anna University Hospital, Brno, Czechia
| | - Yang Cai
- CB Joint MHNCRL, Hospital and School of Stomatology, Guizhou Medical University, Guiyang, China
| | - Ambreen Rehman
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Luke Gammon
- Phenotypic Screening Facility, Blizard Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Usama Sharif Ahmad
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Jutamas Uttagomol
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Eric Kenneth Parkinson
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Farida Fortune
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - Hong Wan
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| |
Collapse
|
16
|
Balmer P, Hariton WVJ, Sayar BS, Jagannathan V, Galichet A, Leeb T, Roosje P, Müller EJ. SUV39H2 epigenetic silencing controls fate conversion of epidermal stem and progenitor cells. J Cell Biol 2021; 220:211810. [PMID: 33604655 PMCID: PMC7898489 DOI: 10.1083/jcb.201908178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/04/2020] [Accepted: 01/21/2021] [Indexed: 12/31/2022] Open
Abstract
Epigenetic histone trimethylation on lysine 9 (H3K9me3) represents a major molecular signal for genome stability and gene silencing conserved from worms to man. However, the functional role of the H3K9 trimethylases SUV39H1/2 in mammalian tissue homeostasis remains largely unknown. Here, we use a spontaneous dog model with monogenic inheritance of a recessive SUV39H2 loss-of-function variant and impaired differentiation in the epidermis, a self-renewing tissue fueled by stem and progenitor cell proliferation and differentiation. Our results demonstrate that SUV39H2 maintains the stem and progenitor cell pool by restricting fate conversion through H3K9me3 repressive marks on gene promoters encoding components of the Wnt/p63/adhesion axis. When SUV39H2 function is lost, repression is relieved, and enhanced Wnt activity causes progenitor cells to prematurely exit the cell cycle, a process mimicked by pharmacological Wnt activation in primary canine, human, and mouse keratinocytes. As a consequence, the stem cell growth potential of cultured SUV39H2-deficient canine keratinocytes is exhausted while epidermal differentiation and genome stability are compromised. Collectively, our data identify SUV39H2 and potentially also SUV39H1 as major gatekeepers in the delicate balance of progenitor fate conversion through H3K9me3 rate-limiting road blocks in basal layer keratinocytes.
Collapse
Affiliation(s)
- Pierre Balmer
- Division of Clinical Dermatology, Department of Clinical Veterinary Medicine, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Bern, Switzerland.,Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - William V J Hariton
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Bern, Switzerland.,Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Beyza S Sayar
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Bern, Switzerland.,Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Vidhya Jagannathan
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Institute of Genetics, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Arnaud Galichet
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Bern, Switzerland.,Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Tosso Leeb
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Institute of Genetics, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Petra Roosje
- Division of Clinical Dermatology, Department of Clinical Veterinary Medicine, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Eliane J Müller
- Dermfocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Department for BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Bern, Switzerland.,Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
17
|
Tham HL, Linder KE, Olivry T. Deep pemphigus (pemphigus vulgaris, pemphigus vegetans and paraneoplastic pemphigus) in dogs, cats and horses: a comprehensive review. BMC Vet Res 2020; 16:457. [PMID: 33228633 PMCID: PMC7686683 DOI: 10.1186/s12917-020-02677-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/13/2020] [Indexed: 12/28/2022] Open
Abstract
Pemphigus is the term used to describe a group of rare mucocutaneous autoimmune bullous diseases characterized by flaccid blisters and erosions of the mucous membranes and/or skin. When the autoantibodies target desmosomes in the deep layers of the epidermis, deep pemphigus variants such as pemphigus vulgaris, pemphigus vegetans and paraneoplastic pemphigus develop. In this article, we will review the signalment, clinical signs, histopathology and treatment outcome of pemphigus vulgaris, pemphigus vegetans and paraneoplastic pemphigus in dogs, cats and horses; where pertinent, we compare the animal diseases to their human homologue. Canine, feline and equine pemphigus vulgaris, pemphigus vegetans and paraneoplastic pemphigus have many features similar to the human counterpart. These chronic and often relapsing autoimmune dermatoses require aggressive immunosuppressive therapy. In animals, the partial-to-complete remission of pemphigus vulgaris and pemphigus vegetans has been achieved with high dose glucocorticoid therapy, with or without adjunct immunosuppressants; the prognosis is grave for paraneoplastic pemphigus.
Collapse
Affiliation(s)
- Heng L. Tham
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA USA
| | - Keith E. Linder
- Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, NC USA
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC USA
| | - Thierry Olivry
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC USA
| |
Collapse
|
18
|
Tong Y, Song Y, Xia C, Deng S. Theoretical and in silico Analyses Reveal MYC as a Dynamic Network Biomarker in Colon and Rectal Cancer. Front Genet 2020; 11:555540. [PMID: 33193630 PMCID: PMC7606845 DOI: 10.3389/fgene.2020.555540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 09/16/2020] [Indexed: 12/24/2022] Open
Abstract
In this article, we make a theoretical and in silico study for uncovering and evaluating biomarkers in colon and rectal cancer (CRC) by the dynamic network biomarker (DNB) theory. We propose a strategy to employ the theoretical concept of UICC TNM classification in CRC. To reveal the critical transition of CRC, the DNB algorithm was implemented to analyze the genome-wide dynamic network through temporal gene expression data. The relationship between gene sets and clinical features was evaluated by weighted gene co-expression network analysis. The results show that MYC was significantly associated with tumor amplification, tumor immune cells, and survival times. The candidate tumor suppressor genes were ZBTB16, MAL, LIFR, and SLIT2. Protein-protein interaction (PPI) analysis shows that these candidate tumor suppressor genes were significant in immune cells. Data from the Human Protein Atlas showed that a high expression of these candidate tumor suppressor genes was associated with favorable prognosis in TNM stages I-IV. In conclusion, this work provides significant and novel information regarding the TNM stage, cause, and consequences of elevated MYC expression in CRC. MYC expression levels had significant negative correlations with tumor suppressor genes and immune cells.
Collapse
Affiliation(s)
- Yanqiu Tong
- Department of Broadcasting and TV, Chongqing Jiaotong University, Chongqing, China
- Laboratory of Forensic Medicine and Biomedical Informatics, Chongqing Medical University, Chongqing, China
| | - Yang Song
- Department of Medical Informatics, Chongqing Medical University, Chongqing, China
| | - Chuanhui Xia
- School of Materials Science and Engineering, Chongqing Jiaotong University, Chongqing, China
| | - Shixiong Deng
- Laboratory of Forensic Medicine and Biomedical Informatics, Chongqing Medical University, Chongqing, China
| |
Collapse
|
19
|
Yang M, Wu H, Zhao M, Chang C, Lu Q. The pathogenesis of bullous skin diseases. J Transl Autoimmun 2019; 2:100014. [PMID: 32743502 PMCID: PMC7388362 DOI: 10.1016/j.jtauto.2019.100014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/14/2019] [Accepted: 08/21/2019] [Indexed: 01/13/2023] Open
Abstract
Bullous skin diseases are a group of dermatoses characterized by blisters and bullae in the skin and mucous membranes. The etiology and pathogenesis of bullous skin diseases are not completely clear. The most common are pemphigus and bullous pemphigoid (BP). Autoantibodies play critical roles in their pathogenesis. Abnormalities in the adhesion between keratinocytes in patients with pemphigus leads to acantholysis and formation of intra-epidermal blisters. Anti-desmoglein autoantibodies are present both in the circulation and skin lesions of patients with pemphigus. The deficient adhesion of keratinocytes to the basement membrane in BP patients gives rise to subepidermal blisters. Autoantibodies against the components of hemidesmosome can be detected in BP patients. Many novel therapeutics based on knowledge of the pathogenesis have emerged in recent years.
Collapse
Affiliation(s)
- Miao Yang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, PR China
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, PR China
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, PR China
| | - Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, 95616, USA
- Division of Pediatric Immunology and Allergy, Joe DiMaggio Children’s Hospital, Hollywood, FL, 33021, USA
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, PR China
| |
Collapse
|
20
|
Ivars M, España A, Alzuguren P, Pelacho B, Lasarte JJ, López-Zabalza MJ. The involvement of ADAM10 in acantholysis in mucocutaneous pemphigus vulgaris depends on the autoantibody profile of each patient. Br J Dermatol 2019; 182:1194-1204. [PMID: 31370093 DOI: 10.1111/bjd.18382] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2019] [Indexed: 01/05/2023]
Abstract
BACKGROUND Acantholysis in pemphigus vulgaris (PV) may be triggered by desmoglein (Dsg) and non-Dsg autoantibodies. The autoantibody profile of each patient results in distinct intracellular signalling patterns. OBJECTIVES Based on our previous findings, we aimed to elucidate whether PV acantholysis in a mouse model may be mediated by activation of a disintegrin and metalloproteinase 10 (ADAM10). METHODS We used three PV-IgG fractions from different patients containing high or low levels of anti-Dsg1 and anti-Dsg3 antibodies, and the presence or not of anti-desmocollin (Dsc) antibodies, using a passive transfer mouse model of PV. RESULTS Although all of the PV-IgG fractions produced suprabasal acantholysis, only those containing anti-Dsg1/3, but not anti-Dsc2/3 antibodies, induced ADAM10 activation in a Src-dependent way, and an increase in the epidermal growth factor (EGF) receptor ligands EGF and betacellulin (BTC). In contrast, the presence of anti-Dsc2/3 antibodies, in addition to anti-Dsg1/3, triggered earlier and ADAM10-independent epidermal detachment, with no increase in EGF and BTC, which was associated with an earlier and more intense acantholysis. CONCLUSIONS All PV-IgG fractions produced suprabasal acantholysis, but our results reveal that depending on the levels of anti-Dsg antibodies or the presence of non-Dsg antibodies, such as anti-Dsc, more severe cell-cell epidermal detachment will occur at different times, and in an ADAM10-dependent manner or not. Acantholysis in these different groups of patients with PV may be a consequence of the activation of specific intracellular mechanisms downstream of Autoantibodies binding to Dsg or non-Dsg proteins, and therefore more specific therapeutic approaches in PV should be used. What's already known about this topic? Suprabasal acantholysis in pemphigus vulgaris (PV) may be triggered by both desmoglein (Dsg) and non-Dsg autoantibodies. The autoantibody profile of each patient is associated with a distinct intracellular signalling pattern. What does this study add? In patients with PV with anti-Dsg3 and anti-Dsg1, but not anti-desmocollin (Dsc)3 antibodies, ADAM10 activation is induced in an Src-dependent way, together with an increase in the epidermal growth factor receptor (EGFR) ligands EGF and betacellulin. The presence of anti-Dsc3 antibodies triggers an earlier and ADAM10-independent acantholysis, without increasing EGFR ligands, and is associated with more severe epidermal detachment. Lower levels of anti-Dsc3 antibodies are associated with less severe acantholysis. What is the translational message? In some patients with PV, the severity and the timing for cell-cell detachment seem to depend on the level of anti-Dsg1/3 antibodies, although other as yet uncharacterized antibodies may also participate. These patients with PV would exhibit inhibition of acantholysis by Src, ADAM10, EGF and EGFR inhibitors. In other patients, the presence of non-Dsg antibodies, such as anti-Dsc2/3, would produce an earlier and more severe ADAM10-independent suprabasal acantholysis.
Collapse
Affiliation(s)
- M Ivars
- Department of Dermatology, University Clinic of Navarra, School of Medicine, Pamplona, Navarra, Spain
| | - A España
- Department of Dermatology, University Clinic of Navarra, School of Medicine, Pamplona, Navarra, Spain.,IdiSNA, Navarra Institute for Health Research, Pamplona, Navarra, Spain
| | - P Alzuguren
- Department of Dermatology, University Clinic of Navarra, School of Medicine, Pamplona, Navarra, Spain
| | - B Pelacho
- IdiSNA, Navarra Institute for Health Research, Pamplona, Navarra, Spain.,Laboratory of Regenerative Medicine, Center for Applied Medical Research, University of Navarra, Pamplona, Navarra, Spain
| | - J J Lasarte
- IdiSNA, Navarra Institute for Health Research, Pamplona, Navarra, Spain.,Center for Applied Medical Research, Program of Immunology and Immunotherapy, University of Navarra, Pamplona, Navarra, Spain
| | - M J López-Zabalza
- Department of Biochemistry and Genetics, University of Navarra, Pamplona, Navarra, Spain
| |
Collapse
|
21
|
Didona D, Maglie R, Eming R, Hertl M. Pemphigus: Current and Future Therapeutic Strategies. Front Immunol 2019; 10:1418. [PMID: 31293582 PMCID: PMC6603181 DOI: 10.3389/fimmu.2019.01418] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 06/05/2019] [Indexed: 12/16/2022] Open
Abstract
Pemphigus encompasses a heterogeneous group of autoimmune blistering diseases, which affect both mucous membranes and the skin. The disease usually runs a chronic-relapsing course, with a potentially devastating impact on the patients' quality of life. Pemphigus pathogenesis is related to IgG autoantibodies targeting various adhesion molecules in the epidermis, including desmoglein (Dsg) 1 and 3, major components of desmosomes. The pathogenic relevance of such autoantibodies has been largely demonstrated experimentally. IgG autoantibody binding to Dsg results in loss of epidermal keratinocyte adhesion, a phenomenon referred to as acantholysis. This in turn causes intra-epidermal blistering and the clinical appearance of flaccid blisters and erosions at involved sites. Since the advent of glucocorticoids, the overall prognosis of pemphigus has largely improved. However, mortality persists elevated, since long-term use of high dose corticosteroids and adjuvant steroid-sparing immunosuppressants portend a high risk of serious adverse events, especially infections. Recently, rituximab, a chimeric anti CD20 monoclonal antibody which induces B-cell depletion, has been shown to improve patients' survival, as early rituximab use results in higher disease remission rates, long term clinical response and faster prednisone tapering compared to conventional immunosuppressive therapies, leading to its approval as a first line therapy in pemphigus. Other anti B-cell therapies targeting B-cell receptor or downstream molecules are currently tried in clinical studies. More intriguingly, a preliminary study in a preclinical mouse model of pemphigus has shown promise regarding future therapeutic application of Chimeric Autoantibody Receptor T-cells engineered using Dsg domains to selectively target autoreactive B-cells. Conversely, previous studies from our group have demonstrated that B-cell depletion in pemphigus resulted in secondary impairment of T-cell function; this may account for the observed long-term remission following B-cell recovery in rituximab treated patients. Likewise, our data support the critical role of Dsg-specific T-cell clones in orchestrating the inflammatory response and B-cell activation in pemphigus. Monitoring autoreactive T-cells in patients may indeed provide further information on the role of these cells, and would be the starting point for designating therapies aimed at restoring the lost immune tolerance against Dsg. The present review focuses on current advances, unmet challenges and future perspectives of pemphigus management.
Collapse
Affiliation(s)
- Dario Didona
- Department of Dermatology and Allergology, Philipps University, Marburg, Germany
| | - Roberto Maglie
- Department of Dermatology and Allergology, Philipps University, Marburg, Germany.,Surgery and Translational Medicine, Section of Dermatology, University of Florence, Florence, Italy.,Section of Dermatology, Departement of Health Sciences, University of Florence, Florence, Italy
| | - Rüdiger Eming
- Department of Dermatology and Allergology, Philipps University, Marburg, Germany
| | - Michael Hertl
- Department of Dermatology and Allergology, Philipps University, Marburg, Germany
| |
Collapse
|
22
|
Luong-Gardiol N, Siddiqui I, Pizzitola I, Jeevan-Raj B, Charmoy M, Huang Y, Irmisch A, Curtet S, Angelov GS, Danilo M, Juilland M, Bornhauser B, Thome M, Hantschel O, Chalandon Y, Cazzaniga G, Bourquin JP, Huelsken J, Held W. γ-Catenin-Dependent Signals Maintain BCR-ABL1 + B Cell Acute Lymphoblastic Leukemia. Cancer Cell 2019; 35:649-663.e10. [PMID: 30991025 DOI: 10.1016/j.ccell.2019.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/29/2019] [Accepted: 03/14/2019] [Indexed: 11/23/2022]
Abstract
The BCR-ABL1 fusion protein is the cause of chronic myeloid leukemia (CML) and of a significant fraction of adult-onset B cell acute lymphoblastic leukemia (B-ALL) cases. Using mouse models and patient-derived samples, we identified an essential role for γ-catenin in the initiation and maintenance of BCR-ABL1+ B-ALL but not CML. The selectivity was explained by a partial γ-catenin dependence of MYC expression together with the susceptibility of B-ALL, but not CML, to reduced MYC levels. MYC and γ-catenin enabled B-ALL maintenance by augmenting BIRC5 and enforced BIRC5 expression overcame γ-catenin loss. Since γ-catenin was dispensable for normal hematopoiesis, these lineage- and disease-specific features of canonical Wnt signaling identified a potential therapeutic target for the treatment of BCR-ABL1+ B-ALL.
Collapse
Affiliation(s)
- Noemie Luong-Gardiol
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Imran Siddiqui
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Irene Pizzitola
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Beena Jeevan-Raj
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Mélanie Charmoy
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Yun Huang
- Department of Pediatric Oncology and Children's Research Centre, University Children's Hospital Zürich, Zürich, Switzerland
| | - Anja Irmisch
- Swiss Institute for Experimental Cancer Research (ISREC), Federal University of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Sara Curtet
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Georgi S Angelov
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Maxime Danilo
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland
| | - Mélanie Juilland
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Beat Bornhauser
- Department of Pediatric Oncology and Children's Research Centre, University Children's Hospital Zürich, Zürich, Switzerland
| | - Margot Thome
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Oliver Hantschel
- Swiss Institute for Experimental Cancer Research (ISREC), Federal University of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Yves Chalandon
- Service d'Hématologie, Hôpitaux Universitaire de Genève, Geneva, Switzerland
| | - Gianni Cazzaniga
- Centro Ricerca Tettamanti, Pediatric Clinic University of Milano-Bicocca, Monza, Italy
| | - Jean-Pierre Bourquin
- Department of Pediatric Oncology and Children's Research Centre, University Children's Hospital Zürich, Zürich, Switzerland
| | - Joerg Huelsken
- Swiss Institute for Experimental Cancer Research (ISREC), Federal University of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Werner Held
- Department of Oncology UNIL CHUV, University of Lausanne, Epalinges, Switzerland.
| |
Collapse
|
23
|
Hariton WVJ, Galichet A, Vanden Berghe T, Overmiller AM, Mahoney MG, Declercq W, Müller EJ. Feasibility study for clinical application of caspase-3 inhibitors in Pemphigus vulgaris. Exp Dermatol 2018; 26:1274-1277. [PMID: 29105150 DOI: 10.1111/exd.13458] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2017] [Indexed: 11/29/2022]
Abstract
The potentially severe side effects of systemic corticosteroids and immunosuppressants used in Pemphigus vulgaris (PV) call for novel therapeutic approaches. In this context, pharmacological inhibition of major pathogenic signalling effectors represents a promising alternative. However, we have also shown that overinhibition of effectors required for epidermal homeostasis can exacerbate PV pathophysiology implicating transepidermal keratinocyte fragility. A feedforward target validation therefore preferentially includes studies on knockout mouse models. We previously reported on successful amelioration of PV blisters following inhibition of non-apoptotic, low-level caspase-3. Here, we use conditional, keratinocyte-specific caspase-3-deficient mice (casp3EKO ) to demonstrate (i) absence of keratinocyte fragility upon injection of the potent Dsg3-specific antibody AK23 and (ii) amelioration of blistering on the background of known signalling effectors. Our results provide the experimental proof of concept justifying translation of the caspase-3 inhibitor approach into PV clinical trials.
Collapse
Affiliation(s)
- William V J Hariton
- Department of BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Bern, Switzerland.,Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland.,DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Arnaud Galichet
- Department of BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Bern, Switzerland.,Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland.,DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Tom Vanden Berghe
- Molecular Signaling and Cell Death Unit, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Andrew M Overmiller
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - My G Mahoney
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Wim Declercq
- Molecular Signaling and Cell Death Unit, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Eliane J Müller
- Department of BioMedical Research, Molecular Dermatology and Stem Cell Research, University of Bern, Bern, Switzerland.,Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland.,DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
24
|
Yulis M, Kusters DHM, Nusrat A. Cadherins: cellular adhesive molecules serving as signalling mediators. J Physiol 2018; 596:3883-3898. [PMID: 29968384 PMCID: PMC6117591 DOI: 10.1113/jp275328] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/03/2018] [Indexed: 01/06/2023] Open
Abstract
The single pass, transmembrane proteins of the cadherin family have been appreciated as important proteins that regulate intercellular adhesion. In addition to this critical function, cadherins contribute to important signalling events that control cellular homeostasis. Many examples exist of classical, desmosomal and atypical cadherins participating in the regulation of signalling events that control homeostatic functions in cells. Much of the work on cadherin mediated signalling focuses on classical cadherins or on specific disease states such as pemphigus vulgaris. Cadherin mediated signalling has been shown to play critical roles during development, in proliferation, apoptosis, disease pathobiology and beyond. It is becoming increasingly clear that cadherins operate through a range of molecular mechanisms. The diversity of pathways and cellular functions regulated by cadherins suggests that we have only scratched the surface in terms of the roles that these versatile proteins play in signalling and cellular function.
Collapse
Affiliation(s)
- Mark Yulis
- Department of PathologyThe University of MichiganAnn ArborMI 48109USA
| | | | - Asma Nusrat
- Department of PathologyThe University of MichiganAnn ArborMI 48109USA
| |
Collapse
|
25
|
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: 75] [Impact Index Per Article: 10.7] [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
|
26
|
Abstract
Pemphigus vulgaris (PV) is a life-threatening disease belonging to the pemphigus group of autoimmune intra-epidermal bullous diseases of the skin and mucosae. The therapeutic management of PV remains challenging and, in some cases, conventional therapy is not adequate to induce clinical remission. The cornerstone of PV treatment remains systemic corticosteroids. Although very effective, long-term corticosteroid administration is characterized by substantial adverse effects. Corticosteroid-sparing adjuvant therapies have been employed in the treatment of PV, aiming to reduce the necessary cumulative dose of corticosteroids. Specifically, immunosuppressive agents such as azathioprine and mycophenolate mofetil are widely used in PV. More recently, high-dose intravenous immunoglobulins, immunoadsorption, and rituximab have been established as additional successful therapeutic options. This review covers both conventional and emerging therapies in PV. In addition, it sheds light on potential future treatment strategies for this disease.
Collapse
Affiliation(s)
- Khalaf Kridin
- Department of Dermatology, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
27
|
Yulis M, Quiros M, Hilgarth R, Parkos CA, Nusrat A. Intracellular Desmoglein-2 cleavage sensitizes epithelial cells to apoptosis in response to pro-inflammatory cytokines. Cell Death Dis 2018; 9:389. [PMID: 29523777 PMCID: PMC5844960 DOI: 10.1038/s41419-018-0380-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/29/2018] [Accepted: 02/01/2018] [Indexed: 12/29/2022]
Abstract
Desmosomal cadherins mediate intercellular adhesion and have also been shown to regulate homeostatic signaling in epithelial cells. We have previously reported that select pro-inflammatory cytokines induce Dsg2 ectodomain cleavage and shedding from intestinal epithelial cells (IECs). Dsg2 extracellular cleaved fragments (Dsg2 ECF) function to induce paracrine pro-proliferative signaling in epithelial cells. In this study, we show that exposure of IECs to pro-inflammatory cytokines interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α) resulted in Dsg2 intracellular cleavage and generation of a ~55 kDa fragment (Dsg2 ICF). Dsg2 intracellular cleavage is mediated by caspase-8 and occurs prior to Dsg2 extracellular cleavage and the execution of apoptosis. Expression of exogenous Dsg2 ICF in model IECs resulted in increased sensitivity to apoptotic stimuli and apoptosis execution. Additionally, expression of the Dsg2 ICF repressed the anti-apoptotic Bcl-2 family member proteins Bcl-XL and Mcl1. Taken together, our findings identify a novel mechanism by which pro-inflammatory mediators induce modification of Dsg2 to activate apoptosis and eliminate damaged cells, while also promoting release of Dsg2 ECF that promotes proliferation of neighboring cells and epithelial barrier recovery.
Collapse
Affiliation(s)
- Mark Yulis
- Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Miguel Quiros
- Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Roland Hilgarth
- Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Charles A Parkos
- Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Asma Nusrat
- Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
28
|
Aktary Z, Alaee M, Pasdar M. Beyond cell-cell adhesion: Plakoglobin and the regulation of tumorigenesis and metastasis. Oncotarget 2018; 8:32270-32291. [PMID: 28416759 PMCID: PMC5458283 DOI: 10.18632/oncotarget.15650] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 12/16/2016] [Indexed: 12/13/2022] Open
Abstract
Plakoglobin (also known as? -catenin) is a member of the Armadillo family of proteins and a paralog of β -catenin. Plakoglobin is a component of both the adherens junctions and desmosomes, and therefore plays a vital role in the regulation of cell-cell adhesion. Similar to β -catenin, plakoglobin is capable of participating in cell signaling in addition to its role in cell-cell adhesion. In this context, β -catenin has a well-documented oncogenic potential as a component of the Wnt signaling pathway. In contrast, while some studies have suggested a tumor promoting activity of plakoglobin in a cell/malignancy specific context, it generally acts as a tumor/metastasis suppressor. How plakoglobin acts as a growth/metastasis inhibitory protein has remained, until recently, unclear. Recent evidence suggests that plakoglobin may suppress tumorigenesis and metastasis by multiple mechanisms, including the suppression of oncogenic signaling, interactions with various proteins involved in tumorigenesis and metastasis, and the regulation of the expression of genes involved in these processes. This review is primarily focused on various mechanisms by which plakoglobin may inhibit tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Zackie Aktary
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Institut Curie, Orsay, France
| | - Mahsa Alaee
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Manijeh Pasdar
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
29
|
Alaee M, Padda A, Mehrabani V, Churchill L, Pasdar M. The physical interaction of p53 and plakoglobin is necessary for their synergistic inhibition of migration and invasion. Oncotarget 2018; 7:26898-915. [PMID: 27058623 PMCID: PMC5042024 DOI: 10.18632/oncotarget.8616] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/14/2016] [Indexed: 01/15/2023] Open
Abstract
Plakoglobin (PG) is a paralog of β-catenin with similar adhesive, but contrasting signalling functions. Although β-catenin has well-known oncogenic function, PG generally acts as a tumor/metastasis suppressor by mechanisms that are just beginning to be deciphered. Previously, we showed that PG interacted with wild type (WT) and a number of mutant p53s, and that its tumor/metastasis suppressor activity may be mediated, at least partially, by this interaction. Here, carcinoma cell lines deficient in both p53 and PG (H1299), or expressing mutant p53 in the absence of PG (SCC9), were transfected with expression constructs encoding WT and different fragments and deletions of p53 and PG, individually or in pairs. Transfectants were characterized for their in vitro growth, migratory and invasive properties and for mapping the interacting domain of p53 and PG. We showed that when coexpressed, p53-WT and PG-WT cooperated to decrease growth, and acted synergistically to significantly reduce cell migration and invasion. The DNA-binding domain of p53 and C-terminal domain of PG mediated p53/PG interaction, and furthermore, the C-terminus of PG played a central role in the inhibition of invasion in association with p53.
Collapse
Affiliation(s)
- Mahsa Alaee
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
| | - Amarjot Padda
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
| | - Vahedah Mehrabani
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
| | - Lucas Churchill
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
| | - Manijeh Pasdar
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
| |
Collapse
|
30
|
Piven OO, Winata CL. The canonical way to make a heart: β-catenin and plakoglobin in heart development and remodeling. Exp Biol Med (Maywood) 2017; 242:1735-1745. [PMID: 28920469 PMCID: PMC5714149 DOI: 10.1177/1535370217732737] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 08/29/2017] [Indexed: 12/12/2022] Open
Abstract
The main mediator of the canonical Wnt pathway, β-catenin, is a major effector of embryonic development, postnatal tissue homeostasis, and adult tissue regeneration. The requirement for β-catenin in cardiogenesis and embryogenesis has been well established. However, many questions regarding the molecular mechanisms by which β-catenin and canonical Wnt signaling regulate these developmental processes remain unanswered. An interesting question that emerged from our studies concerns how β-catenin signaling is modulated through interaction with other factors. Recent experimental data implicate new players in canonical Wnt signaling, particularly those which modulate β-catenin function in many its biological processes, including cardiogenesis. One of the interesting candidates is plakoglobin, a little-studied member of the catenin family which shares several mechanistic and functional features with its close relative, β-catenin. Here we have focused on the function of β-catenin in cardiogenesis. We also summarize findings on plakoglobin signaling function and discuss possible interplays between β-catenin and plakoglobin in the regulation of embryonic heart development. Impact statement Heart development, function, and remodeling are complex processes orchestrated by multiple signaling networks. This review examines our current knowledge of the role of canonical Wnt signaling in cardiogenesis and heart remodeling, focusing primarily on the mechanistic action of its effector β-catenin. We summarize the generally accepted understanding of the field based on experimental in vitro and in vivo data, and address unresolved questions in the field, specifically relating to the role of canonical Wnt signaling in heart maturation and regeneration. What are the modulators of canonical Wnt, and particularly what are the potential roles of plakoglobin, a close relative of β-catenin, in regulating Wnt signaling?Answers to these questions will enhance our understanding of the mechanism by which the canonical Wnt signaling regulates development of the heart and its regeneration after damage.
Collapse
Affiliation(s)
- Oksana O Piven
- Institute of Molecular Biology and Genetic, Kyiv 0314, Ukraine
| | - Cecilia L Winata
- International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
- Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| |
Collapse
|
31
|
Tavakolpour S. Current and future treatment options for pemphigus: Is it time to move towards more effective treatments? Int Immunopharmacol 2017; 53:133-142. [DOI: 10.1016/j.intimp.2017.10.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/18/2017] [Accepted: 10/20/2017] [Indexed: 02/07/2023]
|
32
|
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: 88] [Impact Index Per Article: 11.0] [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
|
33
|
Trop-Steinberg S, Azar Y. Is Myc an Important Biomarker? Myc Expression in Immune Disorders and Cancer. Am J Med Sci 2017; 355:67-75. [PMID: 29289266 DOI: 10.1016/j.amjms.2017.06.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/17/2017] [Accepted: 06/14/2017] [Indexed: 01/08/2023]
Abstract
The proto-oncogene Myc serves as a paradigm for understanding the dynamics of transcriptional regulation. Myc protein has been linked to immune dysfunction, cancer development and neoplastic transformation. We review recent research regarding functions of Myc as an important modulator in immune disorders, postallogeneic hematopoietic stem cell transplantation (HSCT) and several cancers. Myc overexpression has been repeatedly linked to immune disorders and specific cancers, such as myasthenia gravis, psoriasis, pemphigus vulgaris, atherosclerosis, long-term allogeneic survival among HSCT patients, (primary) inflammatory breast cancer, (primary) ovarian carcinoma and hematological malignancies: acute myeloid leukemia, chronic myelogenous leukemia, Hodgkin's lymphoma and diffuse large B-cell lymphoma. However, decreased expression of Myc has been observed in HSCT patients who did not survive. Understanding impaired or inappropriate expression of Myc may present a path for the discovery of new targets for therapeutic applications.
Collapse
Affiliation(s)
- Shivtia Trop-Steinberg
- Faculty of Life and Health Sciences (ST-S), JCT Lev Academic Institute, Jerusalem, Israel.
| | - Yehudit Azar
- Department of Bone Marrow Transplantation (YA), Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
34
|
Ludwig RJ, Vanhoorelbeke K, Leypoldt F, Kaya Z, Bieber K, McLachlan SM, Komorowski L, Luo J, Cabral-Marques O, Hammers CM, Lindstrom JM, Lamprecht P, Fischer A, Riemekasten G, Tersteeg C, Sondermann P, Rapoport B, Wandinger KP, Probst C, El Beidaq A, Schmidt E, Verkman A, Manz RA, Nimmerjahn F. Mechanisms of Autoantibody-Induced Pathology. Front Immunol 2017; 8:603. [PMID: 28620373 PMCID: PMC5449453 DOI: 10.3389/fimmu.2017.00603] [Citation(s) in RCA: 334] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/08/2017] [Indexed: 12/22/2022] Open
Abstract
Autoantibodies are frequently observed in healthy individuals. In a minority of these individuals, they lead to manifestation of autoimmune diseases, such as rheumatoid arthritis or Graves' disease. Overall, more than 2.5% of the population is affected by autoantibody-driven autoimmune disease. Pathways leading to autoantibody-induced pathology greatly differ among different diseases, and autoantibodies directed against the same antigen, depending on the targeted epitope, can have diverse effects. To foster knowledge in autoantibody-induced pathology and to encourage development of urgently needed novel therapeutic strategies, we here categorized autoantibodies according to their effects. According to our algorithm, autoantibodies can be classified into the following categories: (1) mimic receptor stimulation, (2) blocking of neural transmission, (3) induction of altered signaling, triggering uncontrolled (4) microthrombosis, (5) cell lysis, (6) neutrophil activation, and (7) induction of inflammation. These mechanisms in relation to disease, as well as principles of autoantibody generation and detection, are reviewed herein.
Collapse
Affiliation(s)
- Ralf J. Ludwig
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Karen Vanhoorelbeke
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Frank Leypoldt
- Neuroimmunology, Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel, Germany
- Neuroimmunology, Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Lübeck, Germany
- Department of Neurology, University of Kiel, Kiel, Germany
| | - Ziya Kaya
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Katja Bieber
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Sandra M. McLachlan
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Medical Center, UCLA School of Medicine, Los Angeles, CA, United States
| | - Lars Komorowski
- Institute for Experimental Immunology, Affiliated to Euroimmun AG, Lübeck, Germany
| | - Jie Luo
- Department of Neuroscience, University of Pennsylvania Medical School, Philadelphia, PA, United States
| | | | | | - Jon M. Lindstrom
- Department of Neuroscience, University of Pennsylvania Medical School, Philadelphia, PA, United States
| | - Peter Lamprecht
- Department of Rheumatology, University of Lübeck, Lübeck, Germany
| | - Andrea Fischer
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany
| | | | - Claudia Tersteeg
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | | | - Basil Rapoport
- Thyroid Autoimmune Disease Unit, Cedars-Sinai Medical Center, UCLA School of Medicine, Los Angeles, CA, United States
| | - Klaus-Peter Wandinger
- Department of Neurology, Institute of Clinical Chemistry, University Medical-Centre Schleswig-Holstein, Lübeck, Germany
| | - Christian Probst
- Institute for Experimental Immunology, Affiliated to Euroimmun AG, Lübeck, Germany
| | - Asmaa El Beidaq
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Enno Schmidt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Alan Verkman
- Department of Medicine, University of California, San Francisco, CA, United States
- Department of Physiology, University of California, San Francisco, CA, United States
| | - Rudolf A. Manz
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Falk Nimmerjahn
- Department of Biology, Institute of Genetics, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
35
|
Abstract
Pemphigus is a group of IgG-mediated autoimmune diseases of stratified squamous epithelia, such as the skin and oral mucosa, in which acantholysis (the loss of cell adhesion) causes blisters and erosions. Pemphigus has three major subtypes: pemphigus vulgaris, pemphigus foliaceus and paraneoplastic pemphigus. IgG autoantibodies are characteristically raised against desmoglein 1 and desmoglein 3, which are cell-cell adhesion molecules found in desmosomes. The sites of blister formation can be physiologically explained by the anti-desmoglein autoantibody profile and tissue-specific expression pattern of desmoglein isoforms. The pathophysiological roles of T cells and B cells have been characterized in mouse models of pemphigus and patients, revealing insights into the mechanisms of autoimmunity. Diagnosis is based on clinical manifestations and confirmed with histological and immunochemical testing. The current first-line treatment is systemic corticosteroids and adjuvant therapies, including immunosuppressive agents, intravenous immunoglobulin and plasmapheresis. Rituximab, a monoclonal antibody against CD20+ B cells, is a promising therapeutic option that may soon become first-line therapy. Pemphigus is one of the best-characterized human autoimmune diseases and provides an ideal paradigm for both basic and clinical research, especially towards the development of antigen-specific immune suppression treatments for autoimmune diseases.
Collapse
|
36
|
Varga N, Mózes J, Keegan H, White C, Kelly L, Pilkington L, Benczik M, Zsuzsanna S, Sobel G, Koiss R, Babarczi E, Nyíri M, Kovács L, Attila S, Kaltenecker B, Géresi A, Kocsis A, O'Leary J, Martin CM, Jeney C. The Value of a Novel Panel of Cervical Cancer Biomarkers for Triage of HPV Positive Patients and for Detecting Disease Progression. Pathol Oncol Res 2016; 23:295-305. [PMID: 27497597 DOI: 10.1007/s12253-016-0094-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/14/2016] [Indexed: 01/12/2023]
Abstract
In the era of primary vaccination against HPV and at the beginning of the low prevalence of cervical lesions, introduction of screening methods that can distinguish between low- and high-grade lesions is necessary in order to maintain the positive predictive value of screening. This case-control study included 562 women who attended cervical screening or were referred for colposcopy and 140 disease free controls, confirmed by histology and/or cytology. The cases were stratified by age. Using routine exfoliated liquid based cytological samples RT-PCR measurements of biomarker genes, high-risk HPV testing and liquid based cytology were performed and used to evaluate different testing protocols including sets of genes/tests with different test cut-offs for the diagnostic panels. Three new panels of cellular biomarkers for improved triage of hrHPV positive women (diagnostic panel) and for prognostic assessment of CIN lesions were proposed. The diagnostic panel (PIK3AP1, TP63 and DSG3) has the potential to distinguish cytologically normal hrHPV+ women from hrHPV+ women with CIN2+. The prognostic gene panels (KRT78, MUC5AC, BPIFB1 and CXCL13, TP63, DSG3) have the ability to differentiate hrHPV+ CIN1 and carcinoma cases. The diagnostic triage panel showed good likelihood ratios for all age groups. The panel showed age-unrelated performance and even better diagnostic value under age 30, a unique feature among the established cervical triage tests. The prognostic gene-panels demonstrated good discriminatory power and oncogenic, anti-oncogenic grouping of genes. The study highlights the potential for the gene expression panels to be used for diagnostic triage and lesion prognostics in cervical cancer screening.
Collapse
Affiliation(s)
- Norbert Varga
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Johanna Mózes
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Helen Keegan
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Christine White
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Lynne Kelly
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Loretto Pilkington
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Márta Benczik
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Schaff Zsuzsanna
- 2nd Department of Pathology, Semmelweis University, Üllöi út 93, Budapest, 1091, Hungary
| | - Gábor Sobel
- 2nd Department of Obstetrics and Gynecology, Semmelweis University, Üllői út 78/a, Budapest, 1082, Hungary
| | - Róbert Koiss
- Department of Gynecology-Oncology, United Hospital of St. Stephan and Laszlo, Nagyvárad tér 1, Budapest, 1087, Hungary
| | - Edit Babarczi
- Department of Gynecology-Oncology, United Hospital of St. Stephan and Laszlo, Nagyvárad tér 1, Budapest, 1087, Hungary
| | - Miklos Nyíri
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Laura Kovács
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Sebe Attila
- Institute of Pathophysiology, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
| | | | - Adrienn Géresi
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Adrienn Kocsis
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - John O'Leary
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Cara M Martin
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Csaba Jeney
- Department of Medical Microbiology, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary.
| |
Collapse
|
37
|
Alaee M, Danesh G, Pasdar M. Plakoglobin Reduces the in vitro Growth, Migration and Invasion of Ovarian Cancer Cells Expressing N-Cadherin and Mutant p53. PLoS One 2016; 11:e0154323. [PMID: 27144941 PMCID: PMC4856367 DOI: 10.1371/journal.pone.0154323] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/12/2016] [Indexed: 12/20/2022] Open
Abstract
Aberrant expression of cadherins and catenins plays pivotal roles in ovarian cancer development and progression. Plakoglobin (PG, γ-catenin) is a paralog of β-catenin with dual adhesive and signaling functions. While β-catenin has known oncogenic function, PG generally acts as a tumor/metastasis suppressor. We recently showed that PG interacted with p53 and that its growth/metastasis inhibitory function may be mediated by this interaction. Very little is known about the role of PG in ovarian cancer. Here, we investigated the in vitro tumor/metastasis suppressor effects of PG in ovarian cancer cell lines with mutant p53 expression and different cadherin profiles. We showed that the N-cadherin expressing and E-cadherin and PG deficient ES-2 cells were highly migratory and invasive, whereas OV-90 cells that express E-cadherin, PG and very little/no N-cadherin were not. Exogenous expression of PG or E-cadherin or N-cadherin knockdown in ES-2 cells (ES-2-E-cad, ES-2-PG and ES-2-shN-cad) significantly reduced their migration and invasion. Also, PG expression or N-cadherin knockdown significantly decreased ES-2 cells growth. Furthermore, PG interacted with both cadherins and with wild type and mutant p53 in normal ovarian and ES-2-PG cell lines, respectively.
Collapse
Affiliation(s)
- Mahsa Alaee
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
| | - Ghazal Danesh
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
| | - Manijeh Pasdar
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
- * E-mail:
| |
Collapse
|
38
|
Hammers CM, Stanley JR. Mechanisms of Disease: Pemphigus and Bullous Pemphigoid. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 11:175-97. [PMID: 26907530 DOI: 10.1146/annurev-pathol-012615-044313] [Citation(s) in RCA: 219] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pemphigus and bullous pemphigoid are autoantibody-mediated blistering skin diseases. In pemphigus, keratinocytes in epidermis and mucous membranes lose cell-cell adhesion, and in pemphigoid, the basal keratinocytes lose adhesion to the basement membrane. Pemphigus lesions are mediated directly by the autoantibodies, whereas the autoantibodies in pemphigoid fix complement and mediate inflammation. In both diseases, the autoantigens have been cloned and characterized; pemphigus antigens are desmogleins (cell adhesion molecules in desmosomes), and pemphigoid antigens are found in hemidesmosomes (which mediate adhesion to the basement membrane). This knowledge has enabled diagnostic testing for these diseases by enzyme-linked immunosorbent assays and dissection of various pathophysiological mechanisms, including direct inhibition of cell adhesion, antibody-induced internalization of antigen, and cell signaling. Understanding these mechanisms of disease has led to rational targeted therapeutic strategies.
Collapse
Affiliation(s)
- Christoph M Hammers
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania 19104; .,Department of Dermatology, University of Luebeck, D-23562 Luebeck, Germany;
| | - John R Stanley
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania 19104;
| |
Collapse
|
39
|
Di Zenzo G, Amber KT, Sayar BS, Müller EJ, Borradori L. Immune response in pemphigus and beyond: progresses and emerging concepts. Semin Immunopathol 2015; 38:57-74. [DOI: 10.1007/s00281-015-0541-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 10/30/2015] [Indexed: 12/18/2022]
|
40
|
Abstract
Desmosomes represent adhesive, spot-like intercellular junctions that in association with intermediate filaments mechanically link neighboring cells and stabilize tissue architecture. In addition to this structural function, desmosomes also act as signaling platforms involved in the regulation of cell proliferation, differentiation, migration, morphogenesis, and apoptosis. Thus, deregulation of desmosomal proteins has to be considered to contribute to tumorigenesis. Proteolytic fragmentation and downregulation of desmosomal cadherins and plaque proteins by transcriptional or epigenetic mechanisms were observed in different cancer entities suggesting a tumor-suppressive role. However, discrepant data in the literature indicate that context-dependent differences based on alternative intracellular, signal transduction lead to altered outcome. Here, modulation of Wnt/β-catenin signaling by plakoglobin or desmoplakin and of epidermal growth factor receptor signaling appears to be of special relevance. This review summarizes current evidence on how desmosomal proteins participate in carcinogenesis, and depicts the molecular mechanisms involved.
Collapse
Affiliation(s)
- Otmar Huber
- a Institute of Biochemistry II, Jena University Hospital, Friedrich-Schiller-University Jena , Nonnenplan 2-4, 07743 Jena , Germany.,b Center for Sepsis Control and Care, Jena University Hospital , Erlanger Allee 101, 07747 Jena , Germany
| | - Iver Petersen
- c Institute of Pathology, Jena University Hospital, Friedrich-Schiller-University Jena , Ziegelmühlenweg 1, 07743 Jena , Germany
| |
Collapse
|
41
|
Abstract
Desmosomes are cell-cell junctions that mediate adhesion and couple the intermediate filament cytoskeleton to sites of cell-cell contact. This architectural arrangement integrates adhesion and cytoskeletal elements of adjacent cells. The importance of this robust adhesion system is evident in numerous human diseases, both inherited and acquired, which occur when desmosome function is compromised. This review focuses on autoimmune and infectious diseases that impair desmosome function. In addition, we discuss emerging evidence that desmosomal genes are often misregulated in cancer. The emphasis of our discussion is placed on the way in which human diseases can inform our understanding of basic desmosome biology and in turn, the means by which fundamental advances in the cell biology of desmosomes might lead to new treatments for acquired diseases of the desmosome.
Collapse
|
42
|
Luyet C, Schulze K, Sayar BS, Howald D, Müller EJ, Galichet A. Preclinical studies identify non-apoptotic low-level caspase-3 as therapeutic target in pemphigus vulgaris. PLoS One 2015; 10:e0119809. [PMID: 25748204 PMCID: PMC4352034 DOI: 10.1371/journal.pone.0119809] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 02/03/2015] [Indexed: 02/07/2023] Open
Abstract
The majority of pemphigus vulgaris (PV) patients suffer from a live-threatening loss of intercellular adhesion between keratinocytes (acantholysis). The disease is caused by auto-antibodies that bind to desmosomal cadherins desmoglein (Dsg) 3 or Dsg3 and Dsg1 in mucous membranes and skin. A currently unresolved controversy in PV is whether apoptosis is involved in the pathogenic process. The objective of this study was to perform preclinical studies to investigate apoptotic pathway activation in PV pathogenesis with the goal to assess its potential for clinical therapy. For this purpose, we investigated mouse and human skin keratinocyte cultures treated with PV antibodies (the experimental Dsg3 monospecific antibody AK23 or PV patients IgG), PV mouse models (passive transfer of AK23 or PVIgG into adult and neonatal mice) as well as PV patients' biopsies (n=6). A combination of TUNEL assay, analyses of membrane integrity, early apoptotic markers such as cleaved poly-ADP-ribose polymerase (PARP) and the collapse of actin cytoskeleton failed to provide evidence for apoptosis in PV pathogenesis. However, the in vitro and in vivo PV models, allowing to monitor progression of lesion formation, revealed an early, transient and low-level caspase-3 activation. Pharmacological inhibition confirmed the functional implication of caspase-3 in major events in PV such as shedding of Dsg3, keratin retraction, proliferation including c-Myc induction, p38MAPK activation and acantholysis. Together, these data identify low-level caspase-3 activation downstream of disrupted Dsg3 trans- or cis-adhesion as a major event in PV pathogenesis that is non-synonymous with apoptosis and represents, unlike apoptotic components, a promising target for clinical therapy. At a broader level, these results posit that an impairment of adhesive functions in concert with low-level, non-lethal caspase-3 activation can evoke profound cellular changes which may be of relevance for other diseases including cancer.
Collapse
Affiliation(s)
- Camille Luyet
- Molecular Dermatology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Katja Schulze
- Molecular Dermatology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Beyza S. Sayar
- Molecular Dermatology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Denise Howald
- Molecular Dermatology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Eliane J. Müller
- Molecular Dermatology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Arnaud Galichet
- Molecular Dermatology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
43
|
Abstract
The arrival of multicellularity in evolution facilitated cell-cell signaling in conjunction with adhesion. As the ectodomains of cadherins interact with each other directly in trans (as well as in cis), spanning the plasma membrane and associating with multiple other entities, cadherins enable the transduction of "outside-in" or "inside-out" signals. We focus this review on signals that originate from the larger family of cadherins that are inwardly directed to the nucleus, and thus have roles in gene control or nuclear structure-function. The nature of cadherin complexes varies considerably depending on the type of cadherin and its context, and we will address some of these variables for classical cadherins versus other family members. Substantial but still fragmentary progress has been made in understanding the signaling mediators used by varied cadherin complexes to coordinate the state of cell-cell adhesion with gene expression. Evidence that cadherin intracellular binding partners also localize to the nucleus is a major point of interest. In some models, catenins show reduced binding to cadherin cytoplasmic tails favoring their engagement in gene control. When bound, cadherins may serve as stoichiometric competitors of nuclear signals. Cadherins also directly or indirectly affect numerous signaling pathways (e.g., Wnt, receptor tyrosine kinase, Hippo, NFκB, and JAK/STAT), enabling cell-cell contacts to touch upon multiple biological outcomes in embryonic development and tissue homeostasis.
Collapse
Affiliation(s)
- Pierre D McCrea
- Department of Genetics, University of Texas MD Anderson Cancer Center; Program in Genes & Development, Graduate School in Biomedical Sciences, Houston, Texas, USA.
| | - Meghan T Maher
- Department of Biology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Cara J Gottardi
- Cellular and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
44
|
Abstract
Cell-cell adhesions are necessary for structural integrity and barrier formation of the epidermis. Here, we discuss insights from genetic and cell biological studies into the roles of individual cell-cell junctions and their composite proteins in regulating epidermal development and function. In addition to individual adhesive functions, we will discuss emerging ideas on mechanosensation/transduction of junctions in the epidermis, noncanonical roles for adhesion proteins, and crosstalk/interdependencies between the junctional systems. These studies have revealed that cell adhesion proteins are connected to many aspects of tissue physiology including growth control, differentiation, and inflammation.
Collapse
Affiliation(s)
- Kaelyn D Sumigray
- Department of Dermatology, Duke University Medical Center, Durham, North Carolina, USA; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Terry Lechler
- Department of Dermatology, Duke University Medical Center, Durham, North Carolina, USA; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA.
| |
Collapse
|
45
|
Brown L, Wan H. Desmoglein 3: a help or a hindrance in cancer progression? Cancers (Basel) 2015; 7:266-86. [PMID: 25629808 PMCID: PMC4381258 DOI: 10.3390/cancers7010266] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/09/2015] [Accepted: 01/16/2015] [Indexed: 02/07/2023] Open
Abstract
Desmoglein 3 is one of seven desmosomal cadherins that mediate cell-cell adhesion in desmosomes. Desmosomes are the intercellular junctional complexes that anchor the intermediate filaments of adjacent cells and confer strong cell adhesion thus are essential in the maintenance of tissue architecture and structural integrity. Like adherens junctions, desmosomes function as tumour suppressors and are down regulated in the process of epithelial-mesenchymal transition and in tumour cell invasion and metastasis. However, recently several studies have shown that various desmosomal components, including desmoglein 3, are up-regulated in cancer with increased levels of expression correlating with the clinical stage of malignancy, implicating their potentiality to serve as a diagnostic and prognostic marker. Furthermore, in vitro studies have demonstrated that overexpression of desmoglein 3 in cancer cell lines activates several signal pathways that have an impact on cell morphology, adhesion and locomotion. These additional signalling roles of desmoglein 3 may not be associated to its adhesive function in desmosomes but rather function outside of the junctions, acting as a key regulator in the control of actin based cellular processes. This review will discuss recent advances which support the role of desmoglein 3 in cancer progression.
Collapse
Affiliation(s)
- Louise Brown
- Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Center for Clinical and Diagnostic Oral Sciences, Institute of Dentistry, Blizard Building, London E1 2AT, UK.
| | - Hong Wan
- Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Center for Clinical and Diagnostic Oral Sciences, Institute of Dentistry, Blizard Building, London E1 2AT, UK.
| |
Collapse
|
46
|
Li X, Ishii N, Ohata C, Furumura M, Hashimoto T. Signalling pathways in pemphigus vulgaris. Exp Dermatol 2014; 23:155-6. [PMID: 24387643 DOI: 10.1111/exd.12317] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2013] [Indexed: 12/20/2022]
Abstract
Acantholysis in pemphigus vulgaris is induced by binding of autoantibodies to desmoglein 3 (Dsg3). The roles of signalling pathways on development of acantholysis have recently been extensively studied. In the study by Sayar et al., recently published in Exp Dermatol, epidermal growth factor receptor (EGFR) signalling was activated in both in vivo and in vitro pemphigus vulgaris experimental models. However, while EGFR inhibitors suppressed activity of p38 mitogen-activated protein kinase (p38MAPK) linearly, they suppressed activity of c-Myc and acantholysis in a non-linear, V-shaped relationship. These findings indicated complicated interactions among EGFR, p38MAPK and c-Myc in pemphigus vulgaris pathology.
Collapse
Affiliation(s)
- Xiaoguang Li
- Department of Dermatology, Kurume University School of Medicine and Kurume University Institute of Cutaneous Cell Biology, Kurume, Fukuoka, Japan
| | | | | | | | | |
Collapse
|
47
|
|
48
|
Sayar BS, Rüegg S, Schmidt E, Sibilia M, Siffert M, Suter MM, Galichet A, Müller EJ. EGFR inhibitors erlotinib and lapatinib ameliorate epidermal blistering in pemphigus vulgaris in a non-linear, V-shaped relationship. Exp Dermatol 2014; 23:33-8. [PMID: 24279293 DOI: 10.1111/exd.12290] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2013] [Indexed: 12/17/2022]
Abstract
Novel insights into intra-cellular signalling involved in pemphigus vulgaris (PV), an autoimmune blistering disease of skin and mucous membranes, are now revealing new therapeutic approaches such as the chemical inhibition of PV-associated signals in conjunction with standard immunosuppressive therapy. However, extensive inhibition of signalling molecules that are required for normal tissue function and integrity may hamper this approach. Using a neonatal PV mouse model, we demonstrate that epidermal blistering can be prevented in a dose-dependent manner by clinically approved EGFR inhibitors erlotinib and lapatinib, but only up to approximately 50% of normal EGFR activity. At lower EGFR activity, blisters again aggravated and were highly exacerbated in mice with a conditional deletion of EGFR. Statistical analysis of the relation between EGFR activity and the extent of skin blistering revealed the best fit with a non-linear, V-shaped curve with a median break point at 52% EGFR activity (P = 0.0005). Moreover, lapatinib (a dual EGFR/ErbB2 inhibitor) but not erlotinib significantly reduced blistering in the oral cavity, suggesting that signalling mechanisms differ between PV predilection sites. Our results demonstrate that future clinical trials evaluating EGFR/ErbB2 inhibitors in PV patients must select treatment doses that retain a specific level of signal molecule activity. These findings may also be of relevance for cancer patients treated with EGFR inhibitors, for whom skin lesions due to extensive EGFR inhibition represent a major threat.
Collapse
Affiliation(s)
- Beyza S Sayar
- Molecular Dermatology, Institute of Animal Pathology, University of Bern, Bern, Switzerland; DermFocus, University of Bern, Bern, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Müller EJ, Galichet A, Wiener D, Marti E, Drögemüller C, Welle M, Roosje P, Leeb T, Suter MM. Keratinocyte biology and pathology. Vet Dermatol 2014; 25:236-8. [PMID: 25124172 DOI: 10.1111/vde.12158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Eliane J Müller
- DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland; Molecular Dermatology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kitajima Y. 150(th) anniversary series: Desmosomes and autoimmune disease, perspective of dynamic desmosome remodeling and its impairments in pemphigus. ACTA ACUST UNITED AC 2014; 21:269-80. [PMID: 25078507 DOI: 10.3109/15419061.2014.943397] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Desmosomes are the most important intercellular adhering junctions that adhere two adjacent keratinocytes directly with desmosomal cadherins, that is, desmogleins (Dsgs) and desmocollins, forming an epidermal sheet. Recently, two cell-cell adhesion states of desmosomes, that is, "stable hyper-adhesion" and "dynamic weak-adhesion" conditions have been recognized. They are mutually reversible through cell signaling events involving protein kinase C (PKC), Src and epidermal growth factor receptor (EGFR) during Ca(2+)-switching and wound healing. This remodeling is impaired in pemphigus vulgaris (PV, an autoimmune blistering disease), caused by anti-Dsg3 antibodies. The antibody binding to Dsg3 activates PKC, Src and EGFR, linked to generation of dynamic weak-adhesion desmosomes, followed by p38MAPK-mediated endocytosis of Dsg3, resulting in the specific depletion of Dsg3 from desmosomes and acantholysis. A variety of pemphigus outside-in signaling may explain different clinical (non-inflammatory, inflammatory, and necrolytic) types of pemphigus. Pemphigus could be referred to a "desmosome-remodeling disease involving pemphigus IgG-activated outside-in signaling events".
Collapse
Affiliation(s)
- Yasuo Kitajima
- Department of Dermatology, Kizawa Memorial Hospital, Professor Emeritus Gifu University School of Medicine , Minokamo City, Gifu Prefecture , Japan
| |
Collapse
|