1
|
Schmitt T, Huber J, Pircher J, Schmidt E, Waschke J. The impact of signaling pathways on the desmosome ultrastructure in pemphigus. Front Immunol 2025; 15:1497241. [PMID: 39882246 PMCID: PMC11774707 DOI: 10.3389/fimmu.2024.1497241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction The autoantibody-driven disease pemphigus vulgaris (PV) impairs desmosome adhesion in the epidermis. In desmosomes, the pemphigus autoantigens desmoglein 1 (Dsg1) and Dsg3 link adjacent cells. Dsgs are clustered by plaque proteins and linked to the keratin cytoskeleton by desmoplakin (Dp). The aim of this study was to identify the impact of several PV-related signaling pathways on desmosome ultrastructure. Methods STED microscopy, Dispase-based dissociation assay. Results As observed using STED microscopy, pemphigus autoantibodies (PV-IgG) reduced desmosome number, decreased desmosome size, increased plaque distance and thickness and caused loss of adhesion. Decreased desmosome number, increased plaque distance and thickness and loss of adhesion correlate with features found for newly assembled immature desmosomes, observed after Ca2+ depletion and repletion. This was paralleled by plaque asymmetry, keratin filament retraction and fragmentation of Dsg1 and Dsg3 immunostaining. Inhibition of each individual signaling pathway investigated here prevented the loss of adhesion and ameliorated keratin retraction. In addition, inhibition of p38MAPK or PLC completely rescued all parameters of desmosomes ultrastructure and increased desmosome number under basal conditions. In contrast, inhibition of MEK1/2 was only partially protective for desmosome size and plaque thickness, whereas inhibition of Src or increase of cAMP decreased desmosome size but increased the desmosome number even in the presence of PV-IgG. Discussion Alterations of the desmosomal plaque ultrastructure are closely related to loss of adhesion and regulated differently by signaling pathways involved in pemphigus pathogenesis. This insight may allow identification of novel treatment options targeting specific steps of desmosome turn-over in the future.
Collapse
Affiliation(s)
- Thomas Schmitt
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilan-Universität (LMU) Munich, München, Germany
| | - Julia Huber
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilan-Universität (LMU) Munich, München, Germany
| | - Julia Pircher
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilan-Universität (LMU) Munich, München, Germany
| | - Enno Schmidt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilan-Universität (LMU) Munich, München, Germany
| |
Collapse
|
2
|
Outla Z, Prechova M, Korelova K, Gemperle J, Gregor M. Mechanics of cell sheets: plectin as an integrator of cytoskeletal networks. Open Biol 2025; 15:240208. [PMID: 39875099 PMCID: PMC11774597 DOI: 10.1098/rsob.240208] [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: 09/18/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025] Open
Abstract
Epithelia are multicellular sheets that form barriers defining the internal and external environments. The constant stresses acting at this interface require that epithelial sheets are mechanically robust and provide a selective barrier to the hostile exterior. These properties are mediated by cellular junctions which are physically linked with heavily crosslinked cytoskeletal networks. Such hardwiring is facilitated by plakins, a family of giant modular proteins which serve as 'molecular bridges' between different cytoskeletal filaments and multiprotein adhesion complexes. Dysfunction of cytoskeletal crosslinking compromises epithelial biomechanics and structural integrity. Subsequent loss of barrier function leads to disturbed tissue homeostasis and pathological consequences such as skin blistering or intestinal inflammation. In this article, we highlight the importance of the cytolinker protein plectin for the functional organization of epithelial cytoskeletal networks. In particular, we focus on the ability of plectin to act as an integrator of the epithelial cytoarchitecture that defines the biomechanics of the whole tissue. Finally, we also discuss the role of cytoskeletal crosslinking in emerging aspects of epithelial mechanobiology that are critical for the maintenance of epithelial homeostasis.
Collapse
Affiliation(s)
- Zuzana Outla
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Magdalena Prechova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Katerina Korelova
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Jakub Gemperle
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Martin Gregor
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
3
|
Dean WF, Albert RM, Nawara TJ, Ubil M, Beggs RR, Mattheyses AL. Dsg2 ectodomain organization increases throughout desmosome assembly. Cell Adh Migr 2024; 18:1-13. [PMID: 38566311 PMCID: PMC10993919 DOI: 10.1080/19336918.2024.2333366] [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: 04/25/2023] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Desmosomes are intercellular junctions that regulate mechanical integrity in epithelia and cardiac muscle. Dynamic desmosome remodeling is essential for wound healing and development, yet the mechanisms governing junction assembly remain elusive. While we and others have shown that cadherin ectodomains are highly organized, how this ordered architecture emerges during assembly is unknown. Using fluorescence polarization microscopy, we show that desmoglein 2 (Dsg2) ectodomain order gradually increases during 8 h of assembly, coinciding with increasing adhesive strength. In a scratch wound assay, we observed a similar increase in order in desmosomes assembling at the leading edge of migratory cells. Together, our findings indicate that cadherin organization is a hallmark of desmosome maturity and may play a role in conferring adhesive strength.
Collapse
Affiliation(s)
- William F. Dean
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rose M. Albert
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tomasz J. Nawara
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Melanie Ubil
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Reena R. Beggs
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alexa L. Mattheyses
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
4
|
Li H, He Y, Wang Y, Xie L, Wu G, Liu X, Duan X, Zhou K, Ning W. The RhoGAP ARHGAP32 interacts with desmoplakin, and is required for desmosomal organization and assembly. J Cell Sci 2024; 137:jcs261901. [PMID: 39258310 DOI: 10.1242/jcs.261901] [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: 12/13/2023] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
Desmosomes play a crucial role in maintaining tissue barrier integrity, particularly in mechanically stressed tissues. The assembly of desmosomes is regulated by the cytoskeleton and its regulators, and desmosomes also function as a central hub for regulating F-actin. However, the specific mechanisms underlying the crosstalk between desmosomes and F-actin remain unclear. Here, we identified that ARHGAP32, a Rho GTPase-activating protein, is located in desmosomes through its interaction with desmoplakin (DSP) via its GAB2-interacting domain (GAB2-ID). We confirmed that ARHGAP32 is required for desmosomal organization, maturation and length regulation. Notably, loss of ARHGAP32 increased formation of F-actin stress fibers and phosphorylation of the regulatory myosin light chain Myl9 at T18/S19. Inhibition of ROCK activity in ARHGAP32-knockout (KO) cells effectively restored desmosomal organization and the integrity of epithelial cell sheets. Moreover, loss of DSP impaired desmosomal ARHGAP32 location and led to decreased actomyosin contractility. ARHGAP32 with a deletion of the GAB2-ID domain showed enhanced association with RhoA in the cytosol and failed to rescue the desmosomal organization in ARHGAP32-KO cells. Collectively, our study unveils that ARHGAP32 associates with and regulates desmosomes by interacting with DSP. This interaction potentially facilitates the crosstalk between desmosomes and F-actin.
Collapse
Affiliation(s)
- Hua Li
- Center for Life Sciences, Yunnan University, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Kunming, Yunnan 650500, China
| | - Yinzhen He
- Center for Life Sciences, Yunnan University, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Kunming, Yunnan 650500, China
| | - Yan Wang
- Center for Life Sciences, Yunnan University, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Kunming, Yunnan 650500, China
| | - Lin Xie
- Center for Life Sciences, Yunnan University, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Kunming, Yunnan 650500, China
| | - Gangyun Wu
- Center for Life Sciences, Yunnan University, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Kunming, Yunnan 650500, China
| | - Xiayu Liu
- Center for Life Sciences, Yunnan University, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Kunming, Yunnan 650500, China
| | - Xiufen Duan
- Center for Life Sciences, Yunnan University, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Kunming, Yunnan 650500, China
| | - Kaiyao Zhou
- Center for Life Sciences, Yunnan University, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Kunming, Yunnan 650500, China
| | - Wenxiu Ning
- Center for Life Sciences, Yunnan University, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Kunming, Yunnan 650500, China
| |
Collapse
|
5
|
Abstract
Desmosomes are relatives of ancient cadherin-based junctions, which emerged late in evolution to ensure the structural integrity of vertebrate tissues by coupling the intermediate filament cytoskeleton to cell-cell junctions. Their ability to dynamically counter the contractile forces generated by actin-associated adherens junctions is particularly important in tissues under high mechanical stress, such as the skin and heart. Much more than the simple cellular 'spot welds' depicted in textbooks, desmosomes are in fact dynamic structures that can sense and respond to changes in their mechanical environment and external stressors like ultraviolet light and pathogens. These environmental signals are transmitted intracellularly via desmosome-dependent mechanochemical pathways that drive the physiological processes of morphogenesis and differentiation. This Cell Science at a Glance article and the accompanying poster review desmosome structure and assembly, highlight recent insights into how desmosomes integrate chemical and mechanical signaling in the epidermis, and discuss desmosomes as targets in human disease.
Collapse
Affiliation(s)
- Abbey L. Perl
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jenny L. Pokorny
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kathleen J. Green
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
6
|
Rathod M, Franz H, Beyersdorfer V, Wanuske MT, Leal-Fischer K, Hanns P, Stüdle C, Zimmermann A, Buczak K, Schinner C, Spindler V. DPM1 modulates desmosomal adhesion and epidermal differentiation through SERPINB5. J Cell Biol 2024; 223:e202305006. [PMID: 38477878 PMCID: PMC10937187 DOI: 10.1083/jcb.202305006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 11/30/2023] [Accepted: 01/23/2024] [Indexed: 03/14/2024] Open
Abstract
Glycosylation is essential to facilitate cell-cell adhesion and differentiation. We determined the role of the dolichol phosphate mannosyltransferase (DPM) complex, a central regulator for glycosylation, for desmosomal adhesive function and epidermal differentiation. Deletion of the key molecule of the DPM complex, DPM1, in human keratinocytes resulted in weakened cell-cell adhesion, impaired localization of the desmosomal components desmoplakin and desmoglein-2, and led to cytoskeletal organization defects in human keratinocytes. In a 3D organotypic human epidermis model, loss of DPM1 caused impaired differentiation with abnormally increased cornification, reduced thickness of non-corneal layers, and formation of intercellular gaps in the epidermis. Using proteomic approaches, SERPINB5 was identified as a DPM1-dependent interaction partner of desmoplakin. Mechanistically, SERPINB5 reduced desmoplakin phosphorylation at serine 176, which was required for strong intercellular adhesion. These results uncover a novel role of the DPM complex in connecting desmosomal adhesion with epidermal differentiation.
Collapse
Affiliation(s)
- Maitreyi Rathod
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Anatomy and Experimental Morphology, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| | - Henriette Franz
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Vivien Beyersdorfer
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Anatomy and Experimental Morphology, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| | | | | | - Pauline Hanns
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Chiara Stüdle
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Aude Zimmermann
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Katarzyna Buczak
- Proteomics Core Facility, Biocentre, University of Basel, Basel, Switzerland
| | - Camilla Schinner
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Volker Spindler
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Institute of Anatomy and Experimental Morphology, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
7
|
Fülle JB, de Almeida RA, Lawless C, Stockdale L, Yanes B, Lane EB, Garrod DR, Ballestrem C. Proximity Mapping of Desmosomes Reveals a Striking Shift in Their Molecular Neighborhood Associated With Maturation. Mol Cell Proteomics 2024; 23:100735. [PMID: 38342409 PMCID: PMC10943070 DOI: 10.1016/j.mcpro.2024.100735] [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: 05/04/2023] [Revised: 01/29/2024] [Accepted: 02/08/2024] [Indexed: 02/13/2024] Open
Abstract
Desmosomes are multiprotein adhesion complexes that link intermediate filaments to the plasma membrane, ensuring the mechanical integrity of cells across tissues, but how they participate in the wider signaling network to exert their full function is unclear. To investigate this, we carried out protein proximity mapping using biotinylation (BioID). The combined interactomes of the essential desmosomal proteins desmocollin 2a, plakoglobin, and plakophilin 2a (Pkp2a) in Madin-Darby canine kidney epithelial cells were mapped and their differences and commonalities characterized as desmosome matured from Ca2+ dependence to the mature, Ca2+-independent, hyper-adhesive state, which predominates in tissues. Results suggest that individual desmosomal proteins have distinct roles in connecting to cellular signaling pathways and that these roles alter substantially when cells change their adhesion state. The data provide further support for a dualistic concept of desmosomes in which the properties of Pkp2a differ from those of the other, more stable proteins. This body of data provides an invaluable resource for the analysis of desmosome function.
Collapse
Affiliation(s)
- Judith B Fülle
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | | | - Craig Lawless
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Liam Stockdale
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Bian Yanes
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - E Birgitte Lane
- Skin Research Institute of Singapore, Agency of Science Technology and Research (A∗STAR), Singapore, Singapore
| | - David R Garrod
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK.
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK.
| |
Collapse
|
8
|
Roth-Carter QR, Burks HE, Ren Z, Koetsier JL, Tsoi LC, Harms PW, Xing X, Kirma J, Harmon RM, Godsel LM, Perl AL, Gudjonsson JE, Green KJ. Transcriptional profiling of rare acantholytic disorders suggests common mechanisms of pathogenesis. JCI Insight 2023; 8:e168955. [PMID: 37471166 PMCID: PMC10543711 DOI: 10.1172/jci.insight.168955] [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: 01/27/2023] [Accepted: 07/06/2023] [Indexed: 07/22/2023] Open
Abstract
Darier, Hailey-Hailey, and Grover diseases are rare acantholytic skin diseases. While these diseases have different underlying causes, they share defects in cell-cell adhesion in the epidermis and desmosome organization. To better understand the underlying mechanisms leading to disease in these conditions, we performed RNA-seq on lesional skin samples from patients. The transcriptomic profiles of Darier, Hailey-Hailey, and Grover diseases were found to share a remarkable overlap, which did not extend to other common inflammatory skin diseases. Analysis of enriched pathways showed a shared increase in keratinocyte differentiation, and a decrease in cell adhesion and actin organization pathways in Darier, Hailey-Hailey, and Grover diseases. Direct comparison to atopic dermatitis and psoriasis showed that the downregulation in actin organization pathways was a unique feature in the acantholytic skin diseases. Furthermore, upstream regulator analysis suggested that a decrease in SRF/MRTF activity was responsible for the downregulation of actin organization pathways. Staining for MRTFA in lesional skin samples showed a decrease in nuclear MRTFA in patient skin compared with normal skin. These findings highlight the significant level of similarity in the transcriptome of Darier, Hailey-Hailey, and Grover diseases, and identify decreases in actin organization pathways as a unique signature present in these conditions.
Collapse
Affiliation(s)
| | | | - Ziyou Ren
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Lam C. Tsoi
- Department of Dermatology
- Department of Computational Medicine & Bioinformatics
- Department of Biostatistics, and
| | - Paul W. Harms
- Department of Dermatology
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | - Lisa M. Godsel
- Department of Pathology, and
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | | | - Kathleen J. Green
- Department of Pathology, and
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
9
|
Shoykhet M, Dervishi O, Menauer P, Hiermaier M, Moztarzadeh S, Osterloh C, Ludwig RJ, Williams T, Gerull B, Kääb S, Clauss S, Schüttler D, Waschke J, Yeruva S. EGFR inhibition leads to enhanced desmosome assembly and cardiomyocyte cohesion via ROCK activation. JCI Insight 2023; 8:163763. [PMID: 36795511 PMCID: PMC10070108 DOI: 10.1172/jci.insight.163763] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/15/2023] [Indexed: 02/17/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a familial heart disease partly caused by impaired desmosome turnover. Thus, stabilization of desmosome integrity may provide new treatment options. Desmosomes, apart from cellular cohesion, provide the structural framework of a signaling hub. Here, we investigated the role of the epidermal growth factor receptor (EGFR) in cardiomyocyte cohesion. We inhibited EGFR under physiological and pathophysiological conditions using the murine plakoglobin-KO AC model, in which EGFR was upregulated. EGFR inhibition enhanced cardiomyocyte cohesion. Immunoprecipitation showed an interaction of EGFR and desmoglein 2 (DSG2). Immunostaining and atomic force microscopy (AFM) revealed enhanced DSG2 localization and binding at cell borders upon EGFR inhibition. Enhanced area composita length and desmosome assembly were observed upon EGFR inhibition, confirmed by enhanced DSG2 and desmoplakin (DP) recruitment to cell borders. PamGene Kinase assay performed in HL-1 cardiomyocytes treated with erlotinib, an EGFR inhibitor, revealed upregulation of Rho-associated protein kinase (ROCK). Erlotinib-mediated desmosome assembly and cardiomyocyte cohesion were abolished upon ROCK inhibition. Thus, inhibiting EGFR and, thereby, stabilizing desmosome integrity via ROCK might provide treatment options for AC.
Collapse
Affiliation(s)
- Maria Shoykhet
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Orsela Dervishi
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Philipp Menauer
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Matthias Hiermaier
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Sina Moztarzadeh
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Colin Osterloh
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Ralf J Ludwig
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Tatjana Williams
- Comprehensive Heart Failure Center and Department of Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Brenda Gerull
- Comprehensive Heart Failure Center and Department of Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Stefan Kääb
- Medizinische Klinik und Poliklinik I, LMU Hospital, LMU, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modeling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Sebastian Clauss
- Medizinische Klinik und Poliklinik I, LMU Hospital, LMU, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modeling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, LMU Hospital, LMU, Munich, Germany
| | - Dominik Schüttler
- Medizinische Klinik und Poliklinik I, LMU Hospital, LMU, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modeling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, LMU Hospital, LMU, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Sunil Yeruva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| |
Collapse
|
10
|
Nanavati BN, Noordstra I, Verma S, Duszyc K, Green KJ, Yap AS. Desmosome-anchored intermediate filaments facilitate tension-sensitive RhoA signaling for epithelial homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.23.529786. [PMID: 36865131 PMCID: PMC9980054 DOI: 10.1101/2023.02.23.529786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Epithelia are subject to diverse forms of mechanical stress during development and post-embryonic life. They possess multiple mechanisms to preserve tissue integrity against tensile forces, which characteristically involve specialized cell-cell adhesion junctions coupled to the cytoskeleton. Desmosomes connect to intermediate filaments (IF) via desmoplakin (DP)1,2, while the E-cadherin complex links to the actomyosin cytoskeleton in adherens junctions (AJ)3. These distinct adhesion-cytoskeleton systems support different strategies to preserve epithelial integrity, especially against tensile stress. IFs coupled to desmosomes can passively respond to tension by strain-stiffening4-10, whereas for AJs a variety of mechanotransduction mechanisms associated with the E-cadherin apparatus itself11,12, or proximate to the junctions13, can modulate the activity of its associated actomyosin cytoskeleton by cell signaling. We now report a pathway where these systems collaborate for active tension-sensing and epithelial homeostasis. We found that DP was necessary for epithelia to activate RhoA at AJ on tensile stimulation, an effect that required its capacity to couple IF to desmosomes. DP exerted this effect by facilitating the association of Myosin VI with E-cadherin, the mechanosensor for the tension-sensitive RhoA pathway at AJ12. This connection between the DP-IF system and AJ-based tension-sensing promoted epithelial resilience when contractile tension was increased. It further facilitated epithelial homeostasis by allowing apoptotic cells to be eliminated by apical extrusion. Thus, active responses to tensile stress in epithelial monolayers reflect an integrated response of the IF- and actomyosin-based cell-cell adhesion systems.
Collapse
Affiliation(s)
- Bageshri Naimish Nanavati
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland
| | - Ivar Noordstra
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland
| | - Suzie Verma
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland
| | - Kinga Duszyc
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland
| | - Kathleen J. Green
- Departments of Pathology and Dermatology, Feinberg School of Medicine, Northwestern University, Chicago IL 06011 USA
| | - Alpha S. Yap
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland
| |
Collapse
|
11
|
Fuchs M, Radeva MY, Spindler V, Vielmuth F, Kugelmann D, Waschke J. Cytoskeletal anchorage of different Dsg3 pools revealed by combination of hybrid STED/SMFS-AFM. Cell Mol Life Sci 2023; 80:25. [PMID: 36602635 PMCID: PMC9816259 DOI: 10.1007/s00018-022-04681-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023]
Abstract
Desmoglein 3 (Dsg3) is a desmosomal cadherin mediating cell adhesion within desmosomes and is the antigen of the autoimmune blistering skin disease pemphigus vulgaris. Therefore, understanding of the complex desmosome turnover process is of high biomedical relevance. Recently, super resolution microscopy was used to characterize desmosome composition and turnover. However, studies were limited because adhesion measurements on living cells were not possible in parallel. Before desmosomal cadherins are incorporated into nascent desmosomes, they are not bound to intermediate filaments but were suggested to be associated with the actin cytoskeleton. However, direct proof that adhesion of a pool of desmosomal cadherins is dependent on actin is missing. Here, we applied single-molecule force spectroscopy measurements with the novel single molecule hybrid-technique STED/SMFS-AFM to investigate the cytoskeletal anchorage of Dsg3 on living keratinocytes for the first time. By application of pharmacological agents we discriminated two different Dsg3 pools, only one of which is anchored to actin filaments. We applied the actin polymerization inhibitor Latrunculin B to modify the actin cytoskeleton and the PKCα activator PMA to modulate intermediate filament anchorage. On the cellular surface Dsg3 adhesion was actin-dependent. In contrast, at cell-cell contacts, Dsg3 adhesion was independent from actin but rather is regulated by PKC which is well established to control desmosome turn-over via intermediate filament anchorage. Taken together, using the novel STED/SMFS-AFM technique, we demonstrated the existence of two Dsg3 pools with different cytoskeletal anchorage mechanisms.
Collapse
Affiliation(s)
- Michael Fuchs
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Mariya Y Radeva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Volker Spindler
- Department of Biomedicine and Institute of Anatomy, University of Basel, Basel, Switzerland
| | - Franziska Vielmuth
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Daniela Kugelmann
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany.
| |
Collapse
|
12
|
Hegazy M, Koetsier JL, Huffine AL, Broussard JA, Godsel BM, Cohen-Barak E, Sprecher E, Wolfgeher DJ, Kron SJ, Godsel LM, Green KJ. Epidermal stratification requires retromer-mediated desmoglein-1 recycling. Dev Cell 2022; 57:2683-2698.e8. [PMID: 36495876 PMCID: PMC9973369 DOI: 10.1016/j.devcel.2022.11.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 09/12/2022] [Accepted: 11/16/2022] [Indexed: 12/14/2022]
Abstract
Sorting transmembrane cargo is essential for tissue development and homeostasis. However, mechanisms of intracellular trafficking in stratified epidermis are poorly understood. Here, we identify an interaction between the retromer endosomal trafficking component, VPS35, and the desmosomal cadherin, desmoglein-1 (Dsg1). Dsg1 is specifically expressed in stratified epidermis and, when properly localized on the plasma membrane of basal keratinocytes, promotes stratification. We show that the retromer drives Dsg1 recycling from the endo-lysosomal system to the plasma membrane to support human keratinocyte stratification. The retromer-enhancing chaperone, R55, promotes the membrane localization of Dsg1 and a trafficking-deficient mutant associated with a severe inflammatory skin disorder, enhancing its ability to promote stratification. In the absence of Dsg1, retromer association with and expression of the glucose transporter GLUT1 increases, exposing a potential link between Dsg1 deficiency and epidermal metabolism. Our work provides evidence for retromer function in epidermal regeneration, identifying it as a potential therapeutic target.
Collapse
Affiliation(s)
- Marihan Hegazy
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jennifer L Koetsier
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Amber L Huffine
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Joshua A Broussard
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Brendan M Godsel
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Eran Cohen-Barak
- Department of Dermatology, Emek Medical Center, Afula, Israel; Bruce and Ruth Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Eli Sprecher
- Department of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel; Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Donald J Wolfgeher
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL, USA
| | - Stephen J Kron
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL, USA
| | - Lisa M Godsel
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Kathleen J Green
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
13
|
Podyacheva E, Toropova Y. SIRT1 activation and its effect on intercalated disc proteins as a way to reduce doxorubicin cardiotoxicity. Front Pharmacol 2022; 13:1035387. [PMID: 36408244 PMCID: PMC9672938 DOI: 10.3389/fphar.2022.1035387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
According to the World Health Organization, the neoplasm is one of the main reasons for morbidity and mortality worldwide. At the same time, application of cytostatic drugs like an independent type of cancer treatment and in combination with surgical methods, is often associated with the development of cardiovascular complications both in the early and in the delayed period of treatment. Doxorubicin (DOX) is the most commonly used cytotoxic anthracycline antibiotic. DOX can cause both acute and delayed side effects. The problem is still not solved, as evidenced by the continued activity of researchers in terms of developing approaches for the prevention and treatment of cardiovascular complications. It is known, the heart muscle consists of cardiomyocytes connected by intercalated discs (ID), which ensure the structural, electrical, metabolic unity of the heart. Various defects in the ID proteins can lead to the development of cardiovascular diseases of various etiologies, including DOX-induced cardiomyopathy. The search for ways to influence the functioning of ID proteins of the cardiac muscle can become the basis for the creation of new therapeutic approaches to the treatment and prevention of cardiac pathologies. SIRT1 may be an interesting cardioprotective variant due to its wide functional significance. SIRT1 activation triggers nuclear transcription programs that increase the efficiency of cellular, mitochondrial metabolism, increases resistance to oxidative stress, and promotes cell survival. It can be assumed that SIRT1 can not only provide a protective effect at the cardiomyocytes level, leading to an improvement in mitochondrial and metabolic functions, reducing the effects of oxidative stress and inflammatory processes, but also have a protective effect on the functioning of IDs structures of the cardiac muscle.
Collapse
|
14
|
Moch M, Schieren J, Leube RE. Cortical tension regulates desmosomal morphogenesis. Front Cell Dev Biol 2022; 10:946190. [PMID: 36268507 PMCID: PMC9577410 DOI: 10.3389/fcell.2022.946190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 09/14/2022] [Indexed: 12/02/2022] Open
Abstract
Mechanical stability is a fundamental and essential property of epithelial cell sheets. It is in large part determined by cell-cell adhesion sites that are tightly integrated by the cortical cytoskeleton. An intimate crosstalk between the adherens junction-associated contractile actomyosin system and the desmosome-anchored keratin intermediate filament system is decisive for dynamic regulation of epithelial mechanics. A major question in the field is whether and in which way mechanical stress affects junctional plasticity. This is especially true for the desmosome-keratin scaffold whose role in force-sensing is virtually unknown. To examine this question, we inactivated the actomyosin system in human keratinocytes (HaCaT) and canine kidney cells (MDCK) and monitored changes in desmosomal protein turnover. Partial inhibition of myosin II by para-nitro-blebbistatin led to a decrease of the cells' elastic modulus and to reduced desmosomal protein turnover in regions where nascent desmosomes are formed and, to a lower degree, in regions where larger, more mature desmosomes are present. Interestingly, desmosomal proteins are affected differently: a significant decrease in turnover was observed for the desmosomal plaque protein desmoplakin I (DspI), which links keratin filaments to the desmosomal core, and the transmembrane cadherin desmoglein 2 (Dsg2). On the other hand, the turnover of another type of desmosomal cadherin, desmocollin 2 (Dsc2), was not significantly altered under the tested conditions. Similarly, the turnover of the adherens junction-associated E-cadherin was not affected by the low doses of para-nitro-blebbistatin. Inhibition of actin polymerization by low dose latrunculin B treatment and of ROCK-driven actomyosin contractility by Y-27632 treatment also induced a significant decrease in desmosomal DspI turnover. Taken together, we conclude that changes in the cortical force balance affect desmosome formation and growth. Furthermore, they differentially modulate desmosomal protein turnover resulting in changes of desmosome composition. We take the observations as evidence for a hitherto unknown desmosomal mechanosensing and mechanoresponse pathway responding to an altered force balance.
Collapse
|
15
|
Hegazy M, Perl AL, Svoboda SA, Green KJ. Desmosomal Cadherins in Health and Disease. ANNUAL REVIEW OF PATHOLOGY 2022; 17:47-72. [PMID: 34425055 PMCID: PMC8792335 DOI: 10.1146/annurev-pathol-042320-092912] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Desmosomal cadherins are a recent evolutionary innovation that make up the adhesive core of highly specialized intercellular junctions called desmosomes. Desmosomal cadherins, which are grouped into desmogleins and desmocollins, are related to the classical cadherins, but their cytoplasmic domains are tailored for anchoring intermediate filaments instead of actin to sites of cell-cell adhesion. The resulting junctions are critical for resisting mechanical stress in tissues such as the skin and heart. Desmosomal cadherins also act as signaling hubs that promote differentiation and facilitate morphogenesis, creating more complex and effective tissue barriers in vertebrate tissues. Interference with desmosomal cadherin adhesive and supra-adhesive functions leads to a variety of autoimmune, hereditary, toxin-mediated, and malignant diseases. We review our current understanding of how desmosomal cadherins contribute to human health and disease, highlight gaps in our knowledge about their regulation and function, and introduce promising new directions toward combatting desmosome-related diseases.
Collapse
Affiliation(s)
- Marihan Hegazy
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Abbey L. Perl
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Sophia A. Svoboda
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Kathleen J. Green
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA,Department of Dermatology, Feinberg School of Medicine, and Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois 60611, USA
| |
Collapse
|
16
|
Zamani P, Mohammadi H, Mirhoseini SZ. Genome-wide association study and genomic heritabilities for blood protein levels in Lori-Bakhtiari sheep. Sci Rep 2021; 11:23771. [PMID: 34887490 PMCID: PMC8660901 DOI: 10.1038/s41598-021-03290-z] [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: 09/13/2021] [Accepted: 12/01/2021] [Indexed: 01/01/2023] Open
Abstract
Serum protein levels are related to physiological and pathological status of animals and could be affected by both genetic and environmental factors. This study aimed to evaluate genetic variation of serum protein profile in sheep. Blood samples were randomly collected from 96 Lori-Bakhtiari ewes, a heavy meat-type breed. Total protein, albumin, globulin, α1, α2, β and γ globulins and IgG levels were measured in blood serum. The samples were genotyped using the Illumina OvineSNP50 BeadChip. The studied traits adjusted for age, birth type, birth season and estimate of breeding value for body weight were considered as pseudo-phenotypes in genome-wide association analysis. In the GWAS model, the first five principal components were fitted as covariates to correct the biases due to possible population stratification. The Plink, R and GCTA software were used for genome-wide association analysis, construction of Q-Q and Manhattan plots and estimation of genetic variances, respectively. Noticeable genomic heritabilities ± SE were estimated for total and γ globulins (0.868 ± 0.262 and 0.831 ± 0.364, respectively), but other protein fractions had zero or close to zero estimates. Based on the Bonferroni adjusted p values, four QTLs located on 181.7 Mbp of OAR3, 107.7 Mbp of OAR4, 86.3 Mbp of OAR7 and 83.0 Mbp of OAR8 were significantly associated with α1, β, β and γ globulins, respectively. The results showed that the PKP2, IGF2R, SLC22A1 and SLC22A2 genes could be considered as candidate genes for blood serum proteins. The present study showed significant genetic variations of some blood protein fractions.
Collapse
Affiliation(s)
- P Zamani
- Department of Animal Science, Faculty of Agriculture, Bu-Ali Sina University, Hamedan, Iran.
| | - H Mohammadi
- Department of Animal Science, Faculty of Agriculture, Bu-Ali Sina University, Hamedan, Iran
| | - S Z Mirhoseini
- Department of Animal Science, Faculty of Agriculture, University of Guilan, Rasht, Iran
| |
Collapse
|
17
|
Fülle JB, Huppert H, Liebl D, Liu J, Alves de Almeida R, Yanes B, Wright GD, Lane EB, Garrod DR, Ballestrem C. Desmosome dualism - most of the junction is stable, but a plakophilin moiety is persistently dynamic. J Cell Sci 2021; 134:272445. [PMID: 34635908 DOI: 10.1242/jcs.258906] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 10/05/2021] [Indexed: 01/06/2023] Open
Abstract
Desmosomes, strong cell-cell junctions of epithelia and cardiac muscle, link intermediate filaments to cell membranes and mechanically integrate cells across tissues, dissipating mechanical stress. They comprise five major protein classes - desmocollins and desmogleins (the desmosomal cadherins), plakoglobin, plakophilins and desmoplakin - whose individual contribution to the structure and turnover of desmosomes is poorly understood. Using live-cell imaging together with fluorescence recovery after photobleaching (FRAP) and fluorescence loss and localisation after photobleaching (FLAP), we show that desmosomes consist of two contrasting protein moieties or modules: a very stable moiety of desmosomal cadherins, desmoplakin and plakoglobin, and a highly mobile plakophilin (Pkp2a). As desmosomes mature from Ca2+ dependence to Ca2+-independent hyper-adhesion, their stability increases, but Pkp2a remains highly mobile. We show that desmosome downregulation during growth-factor-induced cell scattering proceeds by internalisation of whole desmosomes, which still retain a stable moiety and highly mobile Pkp2a. This molecular mobility of Pkp2a suggests a transient and probably regulatory role for Pkp2a in desmosomes. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Judith B Fülle
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK.,Skin Research Institute of Singapore, Agency of Science Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, 138648 Singapore, Singapore
| | - Henri Huppert
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK.,Institute of Medical Biology, Agency of Science Technology and Research (A*STAR), 61 Biopolis Dr, 138673 Singapore, Singapore
| | - David Liebl
- A*STAR Microscopy Platform, Research Support Centre, Agency of Science Technology and Research (A*STAR), Biopolis 138673 Singapore, Singapore
| | - Jaron Liu
- Institute of Medical Biology, Agency of Science Technology and Research (A*STAR), 61 Biopolis Dr, 138673 Singapore, Singapore
| | - Rogerio Alves de Almeida
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
| | - Bian Yanes
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
| | - Graham D Wright
- Skin Research Institute of Singapore, Agency of Science Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, 138648 Singapore, Singapore.,A*STAR Microscopy Platform, Research Support Centre, Agency of Science Technology and Research (A*STAR), Biopolis 138673 Singapore, Singapore
| | - E Birgitte Lane
- Skin Research Institute of Singapore, Agency of Science Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, 138648 Singapore, Singapore
| | - David R Garrod
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
18
|
Stadiotti I, Di Bona A, Pilato CA, Scalco A, Guarino A, Micheli B, Casella M, Tondo C, Rizzo S, Pilichou K, Thiene G, Frigo AC, Pompilio G, Basso C, Sommariva E, Mongillo M, Zaglia T. Neuropeptide Y promotes adipogenesis of human cardiac mesenchymal stromal cells in arrhythmogenic cardiomyopathy. Int J Cardiol 2021; 342:94-102. [PMID: 34400166 DOI: 10.1016/j.ijcard.2021.08.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/22/2021] [Accepted: 08/06/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Arrhythmogenic Cardiomyopathy (AC) is a familial cardiac disease, mainly caused by mutations in desmosomal genes. AC hearts show fibro-fatty myocardial replacement, which favors stress-related life-threatening arrhythmias, predominantly in the young and athletes. AC lacks effective therapies, as its pathogenesis is poorly understood. Recently, we showed that cardiac Mesenchymal Stromal Cells (cMSCs) contribute to adipose tissue in human AC hearts, although the underlying mechanisms are still unclear. PURPOSE We hypothesize that the sympathetic neurotransmitter, Neuropeptide Y (NPY), participates to cMSC adipogenesis in human AC. METHODS For translation of our findings, we combined in vitro cytochemical, molecular and pharmacologic assays on human cMSCs, from myocardial biopsies of healthy controls and AC patients, with the use of existing drugs to interfere with the predicted AC mechanisms. Sympathetic innervation was inspected in human autoptic heart samples, and NPY plasma levels measured in healthy and AC subjects. RESULTS AC cMSCs expressed higher levels of pro-adipogenic isotypes of NPY-receptors (i.e. Y1-R, Y5-R). Consistently, NPY enhanced adipogenesis in AC cMSCs, which was blocked by FDA-approved Y1-R and Y5-R antagonists. AC-associated PKP2 reduction directly caused NPY-dependent adipogenesis in cMSCs. In support of the involvement of sympathetic neurons (SNs) and NPY in AC myocardial remodeling, patients had elevated NPY plasma levels and, in human AC hearts, SNs accumulated in fatty areas and were close to cMSCs. CONCLUSIONS Independently from the disease origin, AC causes in cMSCs a targetable gain of responsiveness to NPY, which leads to increased adipogenesis, thus playing a role in AC myocardial remodeling.
Collapse
Affiliation(s)
- Ilaria Stadiotti
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Via Parea 4, 20138 Milano, Italy
| | - Anna Di Bona
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy; Veneto Institute of Molecular Medicine, VIMM, via Orus 2, 35129 Padova, Italy
| | - Chiara Assunta Pilato
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Via Parea 4, 20138 Milano, Italy; Department of Biochemical, Surgical and Dentist Sciences, University of Milano, Milano, Italy
| | - Arianna Scalco
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy; Veneto Institute of Molecular Medicine, VIMM, via Orus 2, 35129 Padova, Italy
| | - Anna Guarino
- Cardiovascular Tissue Bank, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138 Milano, Italy
| | - Barbara Micheli
- Cardiovascular Tissue Bank, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138 Milano, Italy
| | - Michela Casella
- Heart Rhythm Center, Department of Clinical Electrophysiology and Cardiac Pacing, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138 Milano, Italy
| | - Claudio Tondo
- Department of Biochemical, Surgical and Dentist Sciences, University of Milano, Milano, Italy; Heart Rhythm Center, Department of Clinical Electrophysiology and Cardiac Pacing, Centro Cardiologico Monzino IRCCS, Via Parea 4, 20138 Milano, Italy
| | - Stefania Rizzo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Kalliopi Pilichou
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Gaetano Thiene
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Anna Chiara Frigo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Via Parea 4, 20138 Milano, Italy; Department of Biochemical, Surgical and Dentist Sciences, University of Milano, Milano, Italy
| | - Cristina Basso
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy
| | - Elena Sommariva
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, Via Parea 4, 20138 Milano, Italy.
| | - Marco Mongillo
- Veneto Institute of Molecular Medicine, VIMM, via Orus 2, 35129 Padova, Italy; Department of Biomedical Science, University of Padova, via Ugo Bassi 58/B, 35131 Padova, Italy; CNR Institute of Neuroscience, Padova, Italy.
| | - Tania Zaglia
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy; Veneto Institute of Molecular Medicine, VIMM, via Orus 2, 35129 Padova, Italy; Department of Biomedical Science, University of Padova, via Ugo Bassi 58/B, 35131 Padova, Italy.
| |
Collapse
|
19
|
Zhang K, Cloonan PE, Sundaram S, Liu F, Das SL, Ewoldt JK, Bays JL, Tomp S, Toepfer CN, Marsiglia JDC, Gorham J, Reichart D, Eyckmans J, Seidman JG, Seidman CE, Chen CS. Plakophilin-2 truncating variants impair cardiac contractility by disrupting sarcomere stability and organization. SCIENCE ADVANCES 2021; 7:eabh3995. [PMID: 34652945 PMCID: PMC8519574 DOI: 10.1126/sciadv.abh3995] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/25/2021] [Indexed: 05/10/2023]
Abstract
Progressive loss of cardiac systolic function in arrhythmogenic cardiomyopathy (ACM) has recently gained attention as an important clinical consideration in managing the disease. However, the mechanisms leading to reduction in cardiac contractility are poorly defined. Here, we use CRISPR gene editing to generate human induced pluripotent stem cells (iPSCs) that harbor plakophilin-2 truncating variants (PKP2tv), the most prevalent ACM-linked mutations. The PKP2tv iPSC–derived cardiomyocytes are shown to have aberrant action potentials and reduced systolic function in cardiac microtissues, recapitulating both the electrical and mechanical pathologies reported in ACM. By combining cell micropatterning with traction force microscopy and live imaging, we found that PKP2tvs impair cardiac tissue contractility by destabilizing cell-cell junctions and in turn disrupting sarcomere stability and organization. These findings highlight the interplay between cell-cell adhesions and sarcomeres required for stabilizing cardiomyocyte structure and function and suggest fundamental pathogenic mechanisms that may be shared among different types of cardiomyopathies.
Collapse
Affiliation(s)
- Kehan Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Paige E. Cloonan
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Subramanian Sundaram
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Feng Liu
- State Key Laboratory of Nonlinear Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
| | - Shoshana L. Das
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jourdan K. Ewoldt
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Jennifer L. Bays
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Samuel Tomp
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Christopher N. Toepfer
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | | | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel Reichart
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Jeroen Eyckmans
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | | | - Christine E. Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Christopher S. Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
20
|
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: 44] [Impact Index Per Article: 11.0] [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
|
21
|
Schmitt T, Waschke J. Autoantibody-Specific Signalling in Pemphigus. Front Med (Lausanne) 2021; 8:701809. [PMID: 34434944 PMCID: PMC8381052 DOI: 10.3389/fmed.2021.701809] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/08/2021] [Indexed: 12/11/2022] Open
Abstract
Pemphigus is a severe autoimmune disease impairing barrier functions of epidermis and mucosa. Autoantibodies primarily target the desmosomal adhesion molecules desmoglein (Dsg) 1 and Dsg 3 and induce loss of desmosomal adhesion. Strikingly, autoantibody profiles in pemphigus correlate with clinical phenotypes. Mucosal-dominant pemphigus vulgaris (PV) is characterised by autoantibodies (PV-IgG) against Dsg3 whereas epidermal blistering in PV and pemphigus foliaceus (PF) is associated with autoantibodies against Dsg1. Therapy in pemphigus is evolving towards specific suppression of autoantibody formation and autoantibody depletion. Nevertheless, during the acute phase and relapses of the disease additional treatment options to stabilise desmosomes and thereby rescue keratinocyte adhesion would be beneficial. Therefore, the mechanisms by which autoantibodies interfere with adhesion of desmosomes need to be characterised in detail. Besides direct inhibition of Dsg adhesion, autoantibodies engage signalling pathways interfering with different steps of desmosome turn-over. With this respect, recent data indicate that autoantibodies induce separate signalling responses in keratinocytes via specific signalling complexes organised by Dsg1 and Dsg3 which transfer the signal of autoantibody binding into the cell. This hypothesis may also explain the different clinical pemphigus phenotypes.
Collapse
Affiliation(s)
- Thomas Schmitt
- Ludwig-Maximilian-Universität München, Anatomische Anstalt, Lehrstuhl Anatomie I - Vegetative Anatomie, Munich, Germany
| | - Jens Waschke
- Ludwig-Maximilian-Universität München, Anatomische Anstalt, Lehrstuhl Anatomie I - Vegetative Anatomie, Munich, Germany
| |
Collapse
|
22
|
Indra I, Troyanovsky RB, Green KJ, Troyanovsky SM. Plakophilin 3 and Par3 facilitate desmosomes' association with the apical junctional complex. Mol Biol Cell 2021; 32:1824-1837. [PMID: 34260281 PMCID: PMC8684708 DOI: 10.1091/mbc.e21-01-0001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Desmosomes (DSMs), together with adherens junctions (AJs) and tight junctions (TJs), constitute the apical cell junctional complex (AJC). While the importance of the apical and basolateral polarity machinery in the organization of AJs and TJs is well established, how DSMs are positioned within the AJC is not understood. Here we use highly polarized DLD1 cells as a model to address how DSMs integrate into the AJC. We found that knockout (KO) of the desmosomal ARM protein Pkp3, but not other major DSM proteins, uncouples DSMs from the AJC without blocking DSM assembly. DLD1 cells also exhibit a prominent extraDSM pool of Pkp3, concentrated in tricellular (tC) contacts. Probing distinct apicobasal polarity pathways revealed that neither the DSM’s association with AJC nor the extraDSM pool of Pkp3 are abolished in cells with defects in Scrib module proteins responsible for basolateral membrane development. However, a loss of the apical polarity protein, Par3, completely eliminates the extraDSM pool of Pkp3 and disrupts AJC localization of desmosomes, dispersing these junctions along the entire length of cell–cell contacts. Our data are consistent with a model whereby Par3 facilitates DSM assembly within the AJC, controlling the availability of an assembly competent pool of Pkp3 stored in tC contacts.
Collapse
Affiliation(s)
| | | | - Kathleen J Green
- Departments of Pathology and Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611
| | | |
Collapse
|
23
|
Jin X, Rosenbohm J, Kim E, Esfahani AM, Seiffert-Sinha K, Wahl JK, Lim JY, Sinha AA, Yang R. Modulation of Mechanical Stress Mitigates Anti-Dsg3 Antibody-Induced Dissociation of Cell-Cell Adhesion. Adv Biol (Weinh) 2021; 5:e2000159. [PMID: 33724731 PMCID: PMC7993752 DOI: 10.1002/adbi.202000159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/21/2020] [Indexed: 12/13/2022]
Abstract
It is becoming increasingly clear that mechanical stress in adhesive junctions plays a significant role in dictating the fate of cell-cell attachment under physiological conditions. Targeted disruption of cell-cell junctions leads to multiple pathological conditions, among them the life-threatening autoimmune blistering disease pemphigus vulgaris (PV). The dissociation of cell-cell junctions by autoantibodies is the hallmark of PV, however, the detailed mechanisms that result in tissue destruction remain unclear. Thus far, research and therapy in PV have focused primarily on immune mechanisms upstream of autoantibody binding, while the biophysical aspects of the cell-cell dissociation process leading to acantholysis are less well studied. In work aimed at illuminating the cellular consequences of autoantibody attachment, it is reported that externally applied mechanical stress mitigates antibody-induced monolayer fragmentation and inhibits p38 MAPK phosphorylation activated by anti-Dsg3 antibody. Further, it is demonstrated that mechanical stress applied externally to cell monolayers enhances cell contractility via RhoA activation and promotes the strengthening of cortical actin, which ultimately mitigates antibody-induced cell-cell dissociation. The study elevates understanding of the mechanism of acantholysis in PV and shifts the paradigm of PV disease development from a focus solely on immune pathways to highlight the key role of physical transformations at the target cell.
Collapse
Affiliation(s)
- Xiaowei Jin
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Jordan Rosenbohm
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Eunju Kim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Amir Monemian Esfahani
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | | | - James K Wahl
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, 68583, USA
| | - Jung Yul Lim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Animesh A Sinha
- Department of Dermatology, University at Buffalo, Buffalo, NY, 14203, USA
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
24
|
Hiermaier M, Kliewe F, Schinner C, Stüdle C, Maly IP, Wanuske MT, Rötzer V, Endlich N, Vielmuth F, Waschke J, Spindler V. The Actin-Binding Protein α-Adducin Modulates Desmosomal Turnover and Plasticity. J Invest Dermatol 2020; 141:1219-1229.e11. [PMID: 33098828 DOI: 10.1016/j.jid.2020.09.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 01/01/2023]
Abstract
Intercellular adhesion is essential for tissue integrity and homeostasis. Desmosomes are abundant in the epidermis and the myocardium-tissues, which are under constantly changing mechanical stresses. Yet, it is largely unclear whether desmosomal adhesion can be rapidly adapted to changing demands, and the mechanisms underlying desmosome turnover are only partially understood. In this study we show that the loss of the actin-binding protein α-adducin resulted in reduced desmosome numbers and prevented the ability of cultured keratinocytes or murine epidermis to withstand mechanical stress. This effect was not primarily caused by decreased levels or impaired adhesive properties of desmosomal molecules but rather by altered desmosome turnover. Mechanistically, reduced cortical actin density in α-adducin knockout keratinocytes resulted in increased mobility of the desmosomal adhesion molecule desmoglein 3 and impaired interactions with E-cadherin, a crucial step in desmosome formation. Accordingly, the loss of α-adducin prevented increased membrane localization of desmoglein 3 in response to cyclic stretch or shear stress. Our data demonstrate the plasticity of desmosomal molecules in response to mechanical stimuli and unravel a mechanism of how the actin cytoskeleton indirectly shapes intercellular adhesion by restricting the membrane mobility of desmosomal molecules.
Collapse
Affiliation(s)
- Matthias Hiermaier
- Department of Biomedicine, University of Basel, Basel, Switzerland; Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Felix Kliewe
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Camilla Schinner
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Chiara Stüdle
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - I Piotr Maly
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marie-Therès Wanuske
- Department of Biomedicine, University of Basel, Basel, Switzerland; Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Vera Rötzer
- Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Franziska Vielmuth
- Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Jens Waschke
- Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Volker Spindler
- Department of Biomedicine, University of Basel, Basel, Switzerland; Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany.
| |
Collapse
|
25
|
Keratin intermediate filaments: intermediaries of epithelial cell migration. Essays Biochem 2020; 63:521-533. [PMID: 31652439 DOI: 10.1042/ebc20190017] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/13/2019] [Accepted: 09/25/2019] [Indexed: 12/12/2022]
Abstract
Migration of epithelial cells is fundamental to multiple developmental processes, epithelial tissue morphogenesis and maintenance, wound healing and metastasis. While migrating epithelial cells utilize the basic acto-myosin based machinery as do other non-epithelial cells, they are distinguished by their copious keratin intermediate filament (KF) cytoskeleton, which comprises differentially expressed members of two large multigene families and presents highly complex patterns of post-translational modification. We will discuss how the unique mechanophysical and biochemical properties conferred by the different keratin isotypes and their modifications serve as finely tunable modulators of epithelial cell migration. We will furthermore argue that KFs together with their associated desmosomal cell-cell junctions and hemidesmosomal cell-extracellular matrix (ECM) adhesions serve as important counterbalances to the contractile acto-myosin apparatus either allowing and optimizing directed cell migration or preventing it. The differential keratin expression in leaders and followers of collectively migrating epithelial cell sheets provides a compelling example of isotype-specific keratin functions. Taken together, we conclude that the expression levels and specific combination of keratins impinge on cell migration by conferring biomechanical properties on any given epithelial cell affecting cytoplasmic viscoelasticity and adhesion to neighboring cells and the ECM.
Collapse
|
26
|
Abstract
Intercalated discs (ICDs) are highly orchestrated structures that connect neighboring cardiomyocytes in the heart. Three major complexes are distinguished in ICD: desmosome, adherens junction (AJ), and gap junction (GJ). Desmosomes are major cell adhesion junctions that anchor cell membrane to the intermediate filament network; AJs connect the actin cytoskeleton of adjacent cells; and gap junctions metabolically and electrically connect the cytoplasm of adjacent cardiomyocytes. All these complexes work as a single unit, the so-called area composita, interdependently rather than individually. Mutation or altered expression of ICD proteins results in various cardiac diseases, such as ARVC (arrhythmogenic right ventricular cardiomyopathy), dilated cardiomyopathy, and hypotrophy cardiomyopathy, eventually leading to heart failure. In this article, we first review the recent findings on the structural organization of ICD and their functions and then focus on the recent advances in molecular pathogenesis of the ICD-related heart diseases, which include two major areas: i) the ICD gene mutations in cardiac diseases, and ii) the involvement of ICD proteins in signal transduction pathways leading to myocardium remodeling and eventual heart failure. These major ICD-related signaling pathways include Wnt/β-catenin pathway, p38 MAPK cascade, Rho-dependent serum response factor (SRF) signaling, calcineurin/NFAT signaling, Hippo kinase cascade, etc., which are differentially regulated in pathological conditions.
Collapse
|
27
|
Badu-Nkansah KA, Lechler T. Proteomic analysis of desmosomes reveals novel components required for epidermal integrity. Mol Biol Cell 2020; 31:1140-1153. [PMID: 32238101 PMCID: PMC7353166 DOI: 10.1091/mbc.e19-09-0542] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Desmosomes are cell–cell adhesions necessary for the maintenance of tissue integrity in the skin and heart. While the core components of desmosomes have been identified, peripheral components that modulate canonical or noncanonical desmosome functions still remain largely unexplored. Here we used targeted proximity labeling approaches to further elaborate the desmosome proteome in epidermal keratinocytes. Quantitative mass spectrometry analysis identified all core desmosomal proteins while uncovering a diverse array of new constituents with broad molecular functions. By individually targeting the inner and outer dense plaques, we defined proteins enriched within these subcompartments. We validated a number of these novel desmosome-associated proteins and find that many are membrane proximal proteins that show a dependence on functional desmosomes for their cortical localization. We further explored the mechanism of localization and function of two novel desmosome-associated adaptor proteins enriched in the desmosome proteome, Crk and Crk-like (CrkL). These proteins interacted with Dsg1 and rely on Dsg1 and desmoplakin for robust cortical localization. Epidermal deletion of both Crk and CrkL resulted in perinatal lethality with defects in desmosome morphology and keratin organization, thus demonstrating the utility of this dataset in identifying novel proteins required for desmosome-dependent epidermal integrity.
Collapse
Affiliation(s)
- Kwabena A Badu-Nkansah
- Department of Dermatology and Department of Cell Biology, Duke University, Durham, NC 27710
| | - Terry Lechler
- Department of Dermatology and Department of Cell Biology, Duke University, Durham, NC 27710
| |
Collapse
|
28
|
Broussard JA, Jaiganesh A, Zarkoob H, Conway DE, Dunn AR, Espinosa HD, Janmey PA, Green KJ. Scaling up single-cell mechanics to multicellular tissues - the role of the intermediate filament-desmosome network. J Cell Sci 2020; 133:jcs228031. [PMID: 32179593 PMCID: PMC7097224 DOI: 10.1242/jcs.228031] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cells and tissues sense, respond to and translate mechanical forces into biochemical signals through mechanotransduction, which governs individual cell responses that drive gene expression, metabolic pathways and cell motility, and determines how cells work together in tissues. Mechanotransduction often depends on cytoskeletal networks and their attachment sites that physically couple cells to each other and to the extracellular matrix. One way that cells associate with each other is through Ca2+-dependent adhesion molecules called cadherins, which mediate cell-cell interactions through adherens junctions, thereby anchoring and organizing the cortical actin cytoskeleton. This actin-based network confers dynamic properties to cell sheets and developing organisms. However, these contractile networks do not work alone but in concert with other cytoarchitectural elements, including a diverse network of intermediate filaments. This Review takes a close look at the intermediate filament network and its associated intercellular junctions, desmosomes. We provide evidence that this system not only ensures tissue integrity, but also cooperates with other networks to create more complex tissues with emerging properties in sensing and responding to increasingly stressful environments. We will also draw attention to how defects in intermediate filament and desmosome networks result in both chronic and acquired diseases.
Collapse
Affiliation(s)
- Joshua A Broussard
- Departments of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Avinash Jaiganesh
- Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Hoda Zarkoob
- Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Daniel E Conway
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Alexander R Dunn
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Horacio D Espinosa
- Department of Mechanical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Paul A Janmey
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathleen J Green
- Departments of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
29
|
Moch M, Schwarz N, Windoffer R, Leube RE. The keratin-desmosome scaffold: pivotal role of desmosomes for keratin network morphogenesis. Cell Mol Life Sci 2020; 77:543-558. [PMID: 31243490 PMCID: PMC7010626 DOI: 10.1007/s00018-019-03198-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/09/2019] [Accepted: 06/17/2019] [Indexed: 11/29/2022]
Abstract
Desmosome-anchored keratin intermediate filaments (KFs) are essential for epithelial coherence. Yet, desmosomal KF attachment and network organization are still unexplored in vivo. We, therefore, monitored KF network morphogenesis in fluorescent keratin 8 knock-in murine embryos revealing keratin enrichment at newly formed desmosomes followed by KF formation, KF elongation and KF fusion. To examine details of this process and its coupling to desmosome formation, we studied fluorescent keratin and desmosomal protein reporter dynamics in the periphery of expanding HaCaT keratinocyte colonies. Less than 3 min after the start of desmosomal proteins clustering non-filamentous keratin enriched at these sites followed by KF formation and elongation. Subsequently, desmosome-anchored KFs merged into stable bundles generating a rim-and-spokes system consisting of subcortical KFs connecting desmosomes to each other and radial KFs connecting desmosomes to the cytoplasmic KF network. We conclude that desmosomes are organizing centers for the KF cytoskeleton with a hitherto unknown nucleation capacity.
Collapse
Affiliation(s)
- Marcin Moch
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Nicole Schwarz
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Reinhard Windoffer
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Rudolf E Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| |
Collapse
|
30
|
Abstract
Arrhythmogenic cardiomyopathy is a genetic disorder characterized by the risk of life-threatening arrhythmias, myocardial dysfunction and fibrofatty replacement of myocardial tissue. Mutations in genes that encode components of desmosomes, the adhesive junctions that connect cardiomyocytes, are the predominant cause of arrhythmogenic cardiomyopathy and can be identified in about half of patients with the condition. However, the molecular mechanisms leading to myocardial destruction, remodelling and arrhythmic predisposition remain poorly understood. Through the development of animal, induced pluripotent stem cell and other models of disease, advances in our understanding of the pathogenic mechanisms of arrhythmogenic cardiomyopathy over the past decade have brought several signalling pathways into focus. These pathways include canonical and non-canonical WNT signalling, the Hippo-Yes-associated protein (YAP) pathway and transforming growth factor-β signalling. These studies have begun to identify potential therapeutic targets whose modulation has shown promise in preclinical models. In this Review, we summarize and discuss the reported molecular mechanisms underlying the pathogenesis of arrhythmogenic cardiomyopathy.
Collapse
|
31
|
Green KJ, Jaiganesh A, Broussard JA. Desmosomes: Essential contributors to an integrated intercellular junction network. F1000Res 2019; 8. [PMID: 31942240 PMCID: PMC6944264 DOI: 10.12688/f1000research.20942.1] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
The development of adhesive connections between cells was critical for the evolution of multicellularity and for organizing cells into complex organs with discrete compartments. Four types of intercellular junction are present in vertebrates: desmosomes, adherens junctions, tight junctions, and gap junctions. All are essential for the development of the embryonic layers and organs as well as adult tissue homeostasis. While each junction type is defined as a distinct entity, it is now clear that they cooperate physically and functionally to create a robust and functionally diverse system. During evolution, desmosomes first appeared in vertebrates as highly specialized regions at the plasma membrane that couple the intermediate filament cytoskeleton at points of strong cell–cell adhesion. Here, we review how desmosomes conferred new mechanical and signaling properties to vertebrate cells and tissues through their interactions with the existing junctional and cytoskeletal network.
Collapse
Affiliation(s)
- Kathleen J Green
- Departments of Pathology and Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Avinash Jaiganesh
- Departments of Pathology and Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Joshua A Broussard
- Departments of Pathology and Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| |
Collapse
|
32
|
Yeh CH, Shen ZQ, Hsiung SY, Wu PC, Teng YC, Chou YJ, Fang SW, Chen CF, Yan YT, Kao LS, Kao CH, Tsai TF. Cisd2 is essential to delaying cardiac aging and to maintaining heart functions. PLoS Biol 2019; 17:e3000508. [PMID: 31593566 PMCID: PMC6799937 DOI: 10.1371/journal.pbio.3000508] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 10/18/2019] [Accepted: 09/24/2019] [Indexed: 11/18/2022] Open
Abstract
CDGSH iron-sulfur domain-containing protein 2 (Cisd2) is pivotal to mitochondrial integrity and intracellular Ca2+ homeostasis. In the heart of Cisd2 knockout mice, Cisd2 deficiency causes intercalated disc defects and leads to degeneration of the mitochondria and sarcomeres, thereby impairing its electromechanical functioning. Furthermore, Cisd2 deficiency disrupts Ca2+ homeostasis via dysregulation of sarco/endoplasmic reticulum Ca2+-ATPase (Serca2a) activity, resulting in an increased level of basal cytosolic Ca2+ and mitochondrial Ca2+ overload in cardiomyocytes. Most strikingly, in Cisd2 transgenic mice, a persistently high level of Cisd2 is sufficient to delay cardiac aging and attenuate age-related structural defects and functional decline. In addition, it results in a younger cardiac transcriptome pattern during old age. Our findings indicate that Cisd2 plays an essential role in cardiac aging and in the heart's electromechanical functioning. They highlight Cisd2 as a novel drug target when developing therapies to delay cardiac aging and ameliorate age-related cardiac dysfunction.
Collapse
Affiliation(s)
- Chi-Hsiao Yeh
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Keelung, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- * E-mail: (C-HY); (T-FT)
| | - Zhao-Qing Shen
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Shao-Yu Hsiung
- Program in Molecular Medicine, School of Life Sciences, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Pei-Chun Wu
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yuan-Chi Teng
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
- Program in Molecular Medicine, School of Life Sciences, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Yi-Ju Chou
- Program in Molecular Medicine, School of Life Sciences, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Su-Wen Fang
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chian-Feng Chen
- Genome Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Ting Yan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Lung-Sen Kao
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Heng Kao
- Center of General Education, Chang Gung University, Taoyuan, Taiwan
| | - Ting-Fen Tsai
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
- Program in Molecular Medicine, School of Life Sciences, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
- Aging and Health Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan
- * E-mail: (C-HY); (T-FT)
| |
Collapse
|
33
|
Calcium as a Key Player in Arrhythmogenic Cardiomyopathy: Adhesion Disorder or Intracellular Alteration? Int J Mol Sci 2019; 20:ijms20163986. [PMID: 31426283 PMCID: PMC6721231 DOI: 10.3390/ijms20163986] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/08/2019] [Accepted: 08/14/2019] [Indexed: 12/20/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited heart disease characterized by sudden death in young people and featured by fibro-adipose myocardium replacement, malignant arrhythmias, and heart failure. To date, no etiological therapies are available. Mutations in desmosomal genes cause abnormal mechanical coupling, trigger pro-apoptotic signaling pathways, and induce fibro-adipose replacement. Here, we discuss the hypothesis that the ACM causative mechanism involves a defect in the expression and/or activity of the cardiac Ca2+ handling machinery, focusing on the available data supporting this hypothesis. The Ca2+ toolkit is heavily remodeled in cardiomyocytes derived from a mouse model of ACM defective of the desmosomal protein plakophilin-2. Furthermore, ACM-related mutations were found in genes encoding for proteins involved in excitation‒contraction coupling, e.g., type 2 ryanodine receptor and phospholamban. As a consequence, the sarcoplasmic reticulum becomes more eager to release Ca2+, thereby inducing delayed afterdepolarizations and impairing cardiac contractility. These data are supported by preliminary observations from patient induced pluripotent stem-cell-derived cardiomyocytes. Assessing the involvement of Ca2+ signaling in the pathogenesis of ACM could be beneficial in the treatment of this life-threatening disease.
Collapse
|
34
|
Kant S, Freytag B, Herzog A, Reich A, Merkel R, Hoffmann B, Krusche CA, Leube RE. Desmoglein 2 mutation provokes skeletal muscle actin expression and accumulation at intercalated discs in murine hearts. J Cell Sci 2019; 132:jcs.199612. [PMID: 30659114 DOI: 10.1242/jcs.199612] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 12/30/2018] [Indexed: 01/05/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is an incurable progressive disease that is linked to mutations in genes coding for components of desmosomal adhesions that are localized to the intercalated disc region, which electromechanically couples adjacent cardiomyocytes. To date, the underlying molecular dysfunctions are not well characterized. In two murine AC models, we find an upregulation of the skeletal muscle actin gene (Acta1), which is known to be a compensatory reaction to compromised heart function. Expression of this gene is elevated prior to visible morphological alterations and clinical symptoms, and persists throughout pathogenesis with an additional major rise during the chronic disease stage. We provide evidence that the increased Acta1 transcription is initiated through nuclear activation of the serum response transcription factor (SRF) by its transcriptional co-activator megakaryoblastic leukemia 1 protein (MKL1, also known as MRTFA). Our data further suggest that perturbed desmosomal adhesion causes Acta1 overexpression during the early stages of the disease, which is amplified by transforming growth factor β (TGFβ) release from fibrotic lesions and surrounding cardiomyocytes during later disease stages. These observations highlight a hitherto unknown molecular AC pathomechanism.
Collapse
Affiliation(s)
- Sebastian Kant
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Benjamin Freytag
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Antonia Herzog
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Anna Reich
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Rudolf Merkel
- Forschungszentrum Jülich, Institute of Complex Systems, ICS-7, Biomechanics, 52428 Jülich, Germany
| | - Bernd Hoffmann
- Forschungszentrum Jülich, Institute of Complex Systems, ICS-7, Biomechanics, 52428 Jülich, Germany
| | - Claudia A Krusche
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Rudolf E Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
35
|
Desmoplakin Harnesses Rho GTPase and p38 Mitogen-Activated Protein Kinase Signaling to Coordinate Cellular Migration. J Invest Dermatol 2018; 139:1227-1236. [PMID: 30579854 DOI: 10.1016/j.jid.2018.11.032] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 02/06/2023]
Abstract
Desmoplakin (DP) is an obligate component of desmosomal cell-cell junctions that links the adhesion plaque to the cytoskeletal intermediate filament network. While a central role for DP in maintaining the structure and stability of the desmosome is well established, recent work has indicated that DP's functions may extend beyond cell-cell adhesion. In our study, we show that loss of DP results in a significant increase in cellular migration, as measured by scratch wound assays, Transwell migration assays, and invasion assays. Loss of DP causes dramatic changes in actin cytoskeleton morphology, including enhanced protrusiveness, and an increase in filopodia length and number. Interestingly, these changes are also observed in single cells, indicating that control of actin morphology is a cell-cell adhesion-independent function of DP. An investigation of cellular signaling pathways uncovered aberrant Rac and p38 mitogen-activated protein kinase (MAPK) activity in DP knockdown cells, restoration of which is sufficient to rescue DP-dependent changes in both cell migration and actin cytoskeleton morphology. Taken together, these data highlight a previously uncharacterized role for the desmosomal cytolinker DP in coordinating cellular migration via p38 MAPK and Rac signaling.
Collapse
|
36
|
Rübsam M, Broussard JA, Wickström SA, Nekrasova O, Green KJ, Niessen CM. Adherens Junctions and Desmosomes Coordinate Mechanics and Signaling to Orchestrate Tissue Morphogenesis and Function: An Evolutionary Perspective. Cold Spring Harb Perspect Biol 2018; 10:a029207. [PMID: 28893859 PMCID: PMC6211388 DOI: 10.1101/cshperspect.a029207] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Cadherin-based adherens junctions (AJs) and desmosomes are crucial to couple intercellular adhesion to the actin or intermediate filament cytoskeletons, respectively. As such, these intercellular junctions are essential to provide not only integrity to epithelia and other tissues but also the mechanical machinery necessary to execute complex morphogenetic and homeostatic intercellular rearrangements. Moreover, these spatially defined junctions serve as signaling hubs that integrate mechanical and chemical pathways to coordinate tissue architecture with behavior. This review takes an evolutionary perspective on how the emergence of these two essential intercellular junctions at key points during the evolution of multicellular animals afforded metazoans with new opportunities to integrate adhesion, cytoskeletal dynamics, and signaling. We discuss known literature on cross-talk between the two junctions and, using the skin epidermis as an example, provide a model for how these two junctions function in concert to orchestrate tissue organization and function.
Collapse
Affiliation(s)
- Matthias Rübsam
- University of Cologne, Department of Dermatology, Cologne Excellence Cluster on Stress Responses in Aging Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC) at the CECAD Research Center, 50931 Cologne, Germany
| | - Joshua A Broussard
- Northwestern University Feinberg School of Medicine, Departments of Pathology and Dermatology, the Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois 60611
| | - Sara A Wickström
- Paul Gerson Unna Group, Skin Homeostasis and Ageing, Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany
| | - Oxana Nekrasova
- Northwestern University Feinberg School of Medicine, Departments of Pathology and Dermatology, the Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois 60611
| | - Kathleen J Green
- Northwestern University Feinberg School of Medicine, Departments of Pathology and Dermatology, the Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois 60611
| | - Carien M Niessen
- University of Cologne, Department of Dermatology, Cologne Excellence Cluster on Stress Responses in Aging Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC) at the CECAD Research Center, 50931 Cologne, Germany
| |
Collapse
|
37
|
Kam CY, Dubash AD, Magistrati E, Polo S, Satchell KJF, Sheikh F, Lampe PD, Green KJ. Desmoplakin maintains gap junctions by inhibiting Ras/MAPK and lysosomal degradation of connexin-43. J Cell Biol 2018; 217:3219-3235. [PMID: 29959233 PMCID: PMC6123000 DOI: 10.1083/jcb.201710161] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 04/26/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022] Open
Abstract
Desmosomal mutations result in potentially deadly cardiocutaneous disease caused by electrical conduction defects and disruption of gap junctions. Kam et al. demonstrate a mechanism whereby loss of the intermediate filament anchoring protein desmoplakin stimulates Cx43 turnover by increasing K-Ras expression, marking Cx43 for lysosomal degradation through ERK1/2 phosphorylation. Desmoplakin (DP) is an obligate component of desmosomes, intercellular adhesive junctions that maintain the integrity of the epidermis and myocardium. Mutations in DP can cause cardiac and cutaneous disease, including arrhythmogenic cardiomyopathy (ACM), an inherited disorder that frequently results in deadly arrhythmias. Conduction defects in ACM are linked to the remodeling and functional interference with Cx43-based gap junctions that electrically and chemically couple cells. How DP loss impairs gap junctions is poorly understood. We show that DP prevents lysosomal-mediated degradation of Cx43. DP loss triggered robust activation of ERK1/2–MAPK and increased phosphorylation of S279/282 of Cx43, which signals clathrin-mediated internalization and subsequent lysosomal degradation of Cx43. RNA sequencing revealed Ras-GTPases as candidates for the aberrant activation of ERK1/2 upon loss of DP. Using a novel Ras inhibitor, Ras/Rap1-specific peptidase (RRSP), or K-Ras knockdown, we demonstrate restoration of Cx43 in DP-deficient cardiomyocytes. Collectively, our results reveal a novel mechanism for the regulation of the Cx43 life cycle by DP in cardiocutaneous models.
Collapse
Affiliation(s)
- Chen Yuan Kam
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Adi D Dubash
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Simona Polo
- Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy.,Dipartimento di Oncologia ed Emato-oncologia, Universita' degli Studi di Milano, Milan, Italy
| | - Karla J F Satchell
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Farah Sheikh
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Paul D Lampe
- Translational Research Program, Public Health Sciences Division, Fred Hutchinson Research Center, Seattle, WA
| | - Kathleen J Green
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL .,Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| |
Collapse
|
38
|
Dorn T, Kornherr J, Parrotta EI, Zawada D, Ayetey H, Santamaria G, Iop L, Mastantuono E, Sinnecker D, Goedel A, Dirschinger RJ, My I, Laue S, Bozoglu T, Baarlink C, Ziegler T, Graf E, Hinkel R, Cuda G, Kääb S, Grace AA, Grosse R, Kupatt C, Meitinger T, Smith AG, Laugwitz KL, Moretti A. Interplay of cell-cell contacts and RhoA/MRTF-A signaling regulates cardiomyocyte identity. EMBO J 2018; 37:e98133. [PMID: 29764980 PMCID: PMC6003642 DOI: 10.15252/embj.201798133] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 03/29/2018] [Accepted: 04/04/2018] [Indexed: 12/13/2022] Open
Abstract
Cell-cell and cell-matrix interactions guide organ development and homeostasis by controlling lineage specification and maintenance, but the underlying molecular principles are largely unknown. Here, we show that in human developing cardiomyocytes cell-cell contacts at the intercalated disk connect to remodeling of the actin cytoskeleton by regulating the RhoA-ROCK signaling to maintain an active MRTF/SRF transcriptional program essential for cardiomyocyte identity. Genetic perturbation of this mechanosensory pathway activates an ectopic fat gene program during cardiomyocyte differentiation, which ultimately primes the cells to switch to the brown/beige adipocyte lineage in response to adipogenesis-inducing signals. We also demonstrate by in vivo fate mapping and clonal analysis of cardiac progenitors that cardiac fat and a subset of cardiac muscle arise from a common precursor expressing Isl1 and Wt1 during heart development, suggesting related mechanisms of determination between the two lineages.
Collapse
Affiliation(s)
- Tatjana Dorn
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Jessica Kornherr
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Elvira I Parrotta
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
- Department of Experimental and Clinical Medicine, Medical School, University of Magna Grecia, Catanzaro, Italy
| | - Dorota Zawada
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Harold Ayetey
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
- Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - Gianluca Santamaria
- Department of Experimental and Clinical Medicine, Medical School, University of Magna Grecia, Catanzaro, Italy
| | - Laura Iop
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Elisa Mastantuono
- Institute of Human Genetics, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Daniel Sinnecker
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
| | - Alexander Goedel
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
| | - Ralf J Dirschinger
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Ilaria My
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Svenja Laue
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Tarik Bozoglu
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | | | - Tilman Ziegler
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Elisabeth Graf
- Institute of Human Genetics, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Rabea Hinkel
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
- IPEK Institute for Cardiovascular Prevention, Klinikum der Universität München - Ludwig-Maximillians-Universität, Munich, Germany
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, Medical School, University of Magna Grecia, Catanzaro, Italy
| | - Stefan Kääb
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München - Ludwig-Maximillians-Universität, Munich, Germany
| | - Andrew A Grace
- Papworth Hospital NHS Foundation Trust, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Robert Grosse
- Pharmacology Institute, Philipps University Marburg, Marburg, Germany
| | - Christian Kupatt
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Austin G Smith
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Karl-Ludwig Laugwitz
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
| | - Alessandra Moretti
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
39
|
Sajda T, Sinha AA. Autoantibody Signaling in Pemphigus Vulgaris: Development of an Integrated Model. Front Immunol 2018; 9:692. [PMID: 29755451 PMCID: PMC5932349 DOI: 10.3389/fimmu.2018.00692] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/21/2018] [Indexed: 01/10/2023] Open
Abstract
Pemphigus vulgaris (PV) is an autoimmune skin blistering disease effecting both cutaneous and mucosal epithelia. Blister formation in PV is known to result from the binding of autoantibodies (autoAbs) to keratinocyte antigens. The primary antigenic targets of pathogenic autoAbs are known to be desmoglein 3, and to a lesser extent, desmoglein 1, cadherin family proteins that partially comprise the desmosome, a protein structure responsible for maintaining cell adhesion, although additional autoAbs, whose role in blister formation is still unclear, are also known to be present in PV patients. Nevertheless, there remain large gaps in knowledge concerning the precise mechanisms through which autoAb binding induces blister formation. Consequently, the primary therapeutic interventions for PV focus on systemic immunosuppression, whose side effects represent a significant health risk to patients. In an effort to identify novel, disease-specific therapeutic targets, a multitude of studies attempting to elucidate the pathogenic mechanisms downstream of autoAb binding, have led to significant advancements in the understanding of autoAb-mediated blister formation. Despite this enhanced characterization of disease processes, a satisfactory explanation of autoAb-induced acantholysis still does not exist. Here, we carefully review the literature investigating the pathogenic disease mechanisms in PV and, taking into account the full scope of results from these studies, provide a novel, comprehensive theory of blister formation in PV.
Collapse
Affiliation(s)
- Thomas Sajda
- Department of Dermatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Animesh A Sinha
- Department of Dermatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
40
|
Spindler V, Waschke J. Pemphigus-A Disease of Desmosome Dysfunction Caused by Multiple Mechanisms. Front Immunol 2018; 9:136. [PMID: 29449846 PMCID: PMC5799217 DOI: 10.3389/fimmu.2018.00136] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/16/2018] [Indexed: 02/01/2023] Open
Abstract
Pemphigus is a severe autoimmune-blistering disease of the skin and mucous membranes caused by autoantibodies reducing desmosomal adhesion between epithelial cells. Autoantibodies against the desmosomal cadherins desmogleins (Dsgs) 1 and 3 as well as desmocollin 3 were shown to be pathogenic, whereas the role of other antibodies is unclear. Dsg3 interactions can be directly reduced by specific autoantibodies. Autoantibodies also alter the activity of signaling pathways, some of which regulate cell cohesion under baseline conditions and alter the turnover of desmosomal components. These pathways include Ca2+, p38MAPK, PKC, Src, EGFR/Erk, and several others. In this review, we delineate the mechanisms relevant for pemphigus pathogenesis based on the histology and the ultrastructure of patients’ lesions. We then dissect the mechanisms which can explain the ultrastructural hallmarks detectable in pemphigus patient skin. Finally, we reevaluate the concept that the spectrum of mechanisms, which induce desmosome dysfunction upon binding of pemphigus autoantibodies, finally defines the clinical phenotype.
Collapse
Affiliation(s)
- Volker Spindler
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Jens Waschke
- Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| |
Collapse
|
41
|
Affiliation(s)
- Nicole A. Najor
- Department of Biology, University of Detroit Mercy, Detroit, Michigan 48221
| |
Collapse
|
42
|
Asrani K, Sood A, Torres A, Georgess D, Phatak P, Kaur H, Dubin A, Talbot CC, Elhelu L, Ewald AJ, Xiao B, Worley P, Lotan TL. mTORC1 loss impairs epidermal adhesion via TGF-β/Rho kinase activation. J Clin Invest 2017; 127:4001-4017. [PMID: 28945203 DOI: 10.1172/jci92893] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 08/02/2017] [Indexed: 12/14/2022] Open
Abstract
Despite its central position in oncogenic intracellular signaling networks, the role of mTORC1 in epithelial development has not been studied extensively in vivo. Here, we have used the epidermis as a model system to elucidate the cellular effects and signaling feedback sequelae of mTORC1 loss of function in epithelial tissue. In mice with conditional epidermal loss of the mTORC1 components Rheb or Rptor, mTORC1 loss of function unexpectedly resulted in a profound skin barrier defect with epidermal abrasions, blistering, and early postnatal lethality, due to a thinned epidermis with decreased desmosomal protein expression and incomplete biochemical differentiation. In mice with mTORC1 loss of function, we found that Rho kinase (ROCK) signaling was constitutively activated, resulting in increased cytoskeletal tension and impaired cell-cell adhesion. Inhibition or silencing of ROCK1 was sufficient to rescue keratinocyte adhesion and biochemical differentiation in these mice. mTORC1 loss of function also resulted in marked feedback upregulation of upstream TGF-β signaling, triggering ROCK activity and its downstream effects on desmosomal gene expression. These findings elucidate a role for mTORC1 in the regulation of epithelial barrier formation, cytoskeletal tension, and cell adhesion, underscoring the complexity of signaling feedback following mTORC1 inhibition.
Collapse
Affiliation(s)
| | | | | | - Dan Georgess
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Pornima Phatak
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, USA
| | | | | | | | | | - Andrew J Ewald
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology, and
| | - Bo Xiao
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Paul Worley
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tamara L Lotan
- Department of Pathology and.,Department of Oncology, and
| |
Collapse
|
43
|
Moncayo-Arlandi J, Brugada R. Unmasking the molecular link between arrhythmogenic cardiomyopathy and Brugada syndrome. Nat Rev Cardiol 2017; 14:744-756. [DOI: 10.1038/nrcardio.2017.103] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
44
|
Hatzfeld M, Keil R, Magin TM. Desmosomes and Intermediate Filaments: Their Consequences for Tissue Mechanics. Cold Spring Harb Perspect Biol 2017; 9:a029157. [PMID: 28096266 PMCID: PMC5453391 DOI: 10.1101/cshperspect.a029157] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Adherens junctions (AJs) and desmosomes connect the actin and keratin filament networks of adjacent cells into a mechanical unit. Whereas AJs function in mechanosensing and in transducing mechanical forces between the plasma membrane and the actomyosin cytoskeleton, desmosomes and intermediate filaments (IFs) provide mechanical stability required to maintain tissue architecture and integrity when the tissues are exposed to mechanical stress. Desmosomes are essential for stable intercellular cohesion, whereas keratins determine cell mechanics but are not involved in generating tension. Here, we summarize the current knowledge of the role of IFs and desmosomes in tissue mechanics and discuss whether the desmosome-keratin scaffold might be actively involved in mechanosensing and in the conversion of chemical signals into mechanical strength.
Collapse
Affiliation(s)
- Mechthild Hatzfeld
- Institute of Molecular Medicine, Division of Pathobiochemistry, Martin-Luther-University Halle-Wittenberg, 06114 Halle, Germany
| | - René Keil
- Institute of Molecular Medicine, Division of Pathobiochemistry, Martin-Luther-University Halle-Wittenberg, 06114 Halle, Germany
| | - Thomas M Magin
- Institute of Biology, Division of Cell and Developmental Biology and Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
45
|
Jones JCR, Kam CY, Harmon RM, Woychek AV, Hopkinson SB, Green KJ. Intermediate Filaments and the Plasma Membrane. Cold Spring Harb Perspect Biol 2017; 9:9/1/a025866. [PMID: 28049646 DOI: 10.1101/cshperspect.a025866] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A variety of intermediate filament (IF) types show intricate association with plasma membrane proteins, including receptors and adhesion molecules. The molecular basis of linkage of IFs to desmosomes at sites of cell-cell interaction and hemidesmosomes at sites of cell-matrix adhesion has been elucidated and involves IF-associated proteins. However, IFs also interact with focal adhesions and cell-surface molecules, including dystroglycan. Through such membrane interactions, it is well accepted that IFs play important roles in the establishment and maintenance of tissue integrity. However, by organizing cell-surface complexes, IFs likely regulate, albeit indirectly, signaling pathways that are key to tissue homeostasis and repair.
Collapse
Affiliation(s)
- Jonathan C R Jones
- The School of Molecular Biosciences, Washington State University, Pullman, Washington 99164
| | - Chen Yuan Kam
- Departments of Dermatology and Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Robert M Harmon
- Departments of Dermatology and Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Alexandra V Woychek
- The School of Molecular Biosciences, Washington State University, Pullman, Washington 99164
| | - Susan B Hopkinson
- The School of Molecular Biosciences, Washington State University, Pullman, Washington 99164
| | - Kathleen J Green
- Departments of Dermatology and Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| |
Collapse
|
46
|
Dubash AD, Kam CY, Aguado BA, Patel DM, Delmar M, Shea LD, Green KJ. Plakophilin-2 loss promotes TGF-β1/p38 MAPK-dependent fibrotic gene expression in cardiomyocytes. J Cell Biol 2016; 212:425-38. [PMID: 26858265 PMCID: PMC4754716 DOI: 10.1083/jcb.201507018] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 01/08/2016] [Indexed: 01/07/2023] Open
Abstract
Members of the desmosome protein family are integral components of the cardiac area composita, a mixed junctional complex responsible for electromechanical coupling between cardiomyocytes. In this study, we provide evidence that loss of the desmosomal armadillo protein Plakophilin-2 (PKP2) in cardiomyocytes elevates transforming growth factor β1 (TGF-β1) and p38 mitogen-activated protein kinase (MAPK) signaling, which together coordinate a transcriptional program that results in increased expression of profibrotic genes. Importantly, we demonstrate that expression of Desmoplakin (DP) is lost upon PKP2 knockdown and that restoration of DP expression rescues the activation of this TGF-β1/p38 MAPK transcriptional cascade. Tissues from PKP2 heterozygous and DP conditional knockout mouse models also exhibit elevated TGF-β1/p38 MAPK signaling and induction of fibrotic gene expression in vivo. These data therefore identify PKP2 and DP as central players in coordination of desmosome-dependent TGF-β1/p38 MAPK signaling in cardiomyocytes, pathways known to play a role in different types of cardiac disease, such as arrhythmogenic or hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Adi D Dubash
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 Department of Biology, Furman University, Greenville SC 29613
| | - Chen Y Kam
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Brian A Aguado
- Department of Biomedical Engineering, Northwestern University, Evanston IL 60208 Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago IL 60611
| | - Dipal M Patel
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Mario Delmar
- New York University School of Medicine, New York, NY 10016
| | - Lonnie D Shea
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208 Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105
| | - Kathleen J Green
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
47
|
Leo-Macias A, Agullo-Pascual E, Delmar M. The cardiac connexome: Non-canonical functions of connexin43 and their role in cardiac arrhythmias. Semin Cell Dev Biol 2015; 50:13-21. [PMID: 26673388 DOI: 10.1016/j.semcdb.2015.12.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 12/01/2015] [Indexed: 12/17/2022]
Abstract
Connexin43 is the major component of gap junctions, an anatomical structure present in the cardiac intercalated disc that provides a low-resistance pathway for direct cell-to-cell passage of electrical charge. Recent studies have shown that in addition to its well-established function as an integral membrane protein that oligomerizes to form gap junctions, Cx43 plays other roles that are independent of channel (or perhaps even hemi-channel) formation. This article discusses non-canonical functions of Cx43. In particular, we focus on the role of Cx43 as a part of a protein interacting network, a connexome, where molecules classically defined as belonging to the mechanical junctions, the gap junctions and the sodium channel complex, multitask and work together to bring about excitability, electrical and mechanical coupling between cardiac cells. Overall, viewing Cx43 as a multi-functional protein, beyond gap junctions, opens a window to better understand the function of the intercalated disc and the pathological consequences that may result from changes in the abundance or localization of Cx43 in the intercalated disc subdomain.
Collapse
Affiliation(s)
- Alejandra Leo-Macias
- The Leon H Charney Division of Cardiology, New York University School of Medicine, New York, NY, United States
| | - Esperanza Agullo-Pascual
- The Leon H Charney Division of Cardiology, New York University School of Medicine, New York, NY, United States
| | - Mario Delmar
- The Leon H Charney Division of Cardiology, New York University School of Medicine, New York, NY, United States.
| |
Collapse
|
48
|
Ellawindy A, Satoh K, Sunamura S, Kikuchi N, Suzuki K, Minami T, Ikeda S, Tanaka S, Shimizu T, Enkhjargal B, Miyata S, Taguchi Y, Handoh T, Kobayashi K, Kobayashi K, Nakayama K, Miura M, Shimokawa H. Rho-Kinase Inhibition During Early Cardiac Development Causes Arrhythmogenic Right Ventricular Cardiomyopathy in Mice. Arterioscler Thromb Vasc Biol 2015; 35:2172-84. [PMID: 26315406 DOI: 10.1161/atvbaha.115.305872] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 08/17/2015] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Arrhythmogenic right ventricular cardiomyopathy (ARVC) is characterized by fibrofatty changes of the right ventricle, ventricular arrhythmias, and sudden death. Though ARVC is currently regarded as a disease of the desmosome, desmosomal gene mutations have been identified only in half of ARVC patients, suggesting the involvement of other associated mechanisms. Rho-kinase signaling is involved in the regulation of intracellular transport and organizes cytoskeletal filaments, which supports desmosomal protein complex at the myocardial cell-cell junctions. Here, we explored whether inhibition of Rho-kinase signaling is involved in the pathogenesis of ARVC. APPROACH AND RESULTS Using 2 novel mouse models with SM22α- or αMHC-restricted overexpression of dominant-negative Rho-kinase, we show that mice with Rho-kinase inhibition in the developing heart (SM22α-restricted) spontaneously develop cardiac dilatation and dysfunction, myocardial fibrofatty changes, and ventricular arrhythmias, resulting in premature sudden death, phenotypes fulfilling the criteria of ARVC in humans. Rho-kinase inhibition in the developing heart results in the development of ARVC phenotypes in dominant-negative Rho-kinase mice through 3 mechanisms: (1) reduction of cardiac cell proliferation and ventricular wall thickness, (2) stimulation of the expression of the proadipogenic noncanonical Wnt ligand, Wnt5b, and the major adipogenic transcription factor, PPARγ (peroxisome proliferator activated receptor-γ), and inhibition of Wnt/β-catenin signaling, and (3) development of desmosomal abnormalities. These mechanisms lead to the development of cardiac dilatation and dysfunction, myocardial fibrofatty changes, and ventricular arrhythmias, ultimately resulting in sudden premature death in this ARVC mouse model. CONCLUSIONS This study demonstrates a novel crucial role of Rho-kinase inhibition during cardiac development in the pathogenesis of ARVC in mice.
Collapse
Affiliation(s)
- Alia Ellawindy
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Kimio Satoh
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Shinichiro Sunamura
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Nobuhiro Kikuchi
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Kota Suzuki
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Tatsuro Minami
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Shohei Ikeda
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Shinichi Tanaka
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Toru Shimizu
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Budbazar Enkhjargal
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Satoshi Miyata
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Yuhto Taguchi
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Tetsuya Handoh
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Kenta Kobayashi
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Kazuto Kobayashi
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Keiko Nakayama
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Masahito Miura
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.)
| | - Hiroaki Shimokawa
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (A.E., K.S., S.S., N.K., K.S., T.M., S.I., S.T., T.S., B.E., S.M., H.S.); and Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan (T.M., S.T.); Department of Clinical Physiology, Health Science, Tohoku University Graduate School of Medicine, Sendai, Japan (Y.T., T.H., M.M.); Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan (K.K., K.K.); and United Centers for Advanced Research and Translational Medicine, Core Center of Cancer Research, Division of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Japan (K.N.).
| |
Collapse
|
49
|
Wang L, Liu S, Zhang H, Hu S, Wei Y. RhoA activity increased in myocardium of arrhythmogenic cardiomyopathy patients and affected connexin 43 protein expression in HL-1 cells. Int J Clin Exp Med 2015; 8:12906-12913. [PMID: 26550207 PMCID: PMC4612892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 07/28/2015] [Indexed: 06/05/2023]
Abstract
Arrhythmogenic cardiomyopathy (AC) is a familial heart muscle disease with mutations of desmosomal gene and its pathogenesis is related with mutations of desmosomal gene and abnormality of connexin43 (Cx43). One of Rho GTPase, RhoA involves in many pathological processes and is regulated by desmosomal gene PKP2. We aim to identify if PKP2 regulate RhoA activity in myocardium of AC patients, the activity change of RhoA in patients' myocardium and to investigate the effect of active RhoA on the protein expression of Cx43 in myocardial cells. The protein expression level was assessed by western blot and the activity of RhoA was assessed by RhoA protein activation assay. Our results showed that the expression of PKP2 was decreased in myocardium of three patients, one of which carried PKP2 mutations. The activity of RhoA in myocardium was increased in myocardium of AC as compared with healthy control except for the patient with PKP2 mutation, the expression of Cx43 was also increased in those patients with increased activity of RhoA. The results in vitro demonstrated that the increase of active RhoA can cause the change of protein expression of Cx43 in HL-1 cardiomyocytes. In conclusion, regulation of RhoA activity is complex in the myocardium of AC and the activity of RhoA is increased in AC patients without PKP2 mutations. What's more, the active RhoA affects the protein expression of Cx43 in vivo and in vitro, this may be the possible disease mechanism of AC.
Collapse
Affiliation(s)
- Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100037, People's Republic of China
| | - Shenghua Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100037, People's Republic of China
| | - Hongliang Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100037, People's Republic of China
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100037, People's Republic of China
| | - Yingjie Wei
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100037, People's Republic of China
| |
Collapse
|
50
|
Fujiwara M, Nagatomo A, Tsuda M, Obata S, Sakuma T, Yamamoto T, Suzuki ST. Desmocollin-2 alone forms functional desmosomal plaques, with the plaque formation requiring the juxtamembrane region and plakophilins. J Biochem 2015; 158:339-53. [DOI: 10.1093/jb/mvv048] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 04/09/2015] [Indexed: 01/04/2023] Open
|