1
|
Badowski C, Benny P, Verma CS, Lane EB. Desmoplakin CSM models unravel mechanisms regulating the binding to intermediate filaments and putative therapeutics for cardiocutaneous diseases. Sci Rep 2024; 14:23206. [PMID: 39369039 PMCID: PMC11455855 DOI: 10.1038/s41598-024-73705-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/19/2024] [Indexed: 10/07/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a common cause of sudden cardiac arrest and death in young adults. It can be induced by different types of mutations throughout the desmoplakin gene including the R2834H mutation in the extreme carboxyterminus tail of desmoplakin (DP CT) which remains structurally uncharacterized and poorly understood. Here, we have created 3D models of DP CT which show the structural effects of AC-inducing mutations as well as the implications of post-translational modifications (PTMs). Our results suggest that, in absence of PTMs, positively charged wildtype DP CT likely folds back onto negatively-charged plectin repeat 14 of nearby plakin repeat domain C (PRD C) contributing to the recruitment of intermediate filaments (IFs). When phosphorylated and methylated, negatively-charged wildtype DP CT would then fold back onto positively-charged plectin repeat 17 of PRD C, promoting the repulsion of intermediate filaments. However, by preventing PTMs, the R2834H mutation would lead to the formation of a cytoplasmic mutant desmoplakin with a constitutively positive DP CT tail that would be aberrantly recruited by cytoplasmic IFs instead of desmosomes, potentially weakening cell-cell contacts and promoting AC. Virtual screening of FDA-approved drug libraries identified several promising drug candidates for the treatment of cardiocutaneous diseases through drug repurposing.
Collapse
Affiliation(s)
- Cedric Badowski
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore.
| | - Paula Benny
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
- Department of Obstetrics and Gynaecology, National University Hospital, National University of Singapore, 1E Kent Ridge Rd, Level 12 NUHS Tower Block, Singapore, 119228, Singapore
- NUS Bia-Echo Asia Centre of Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Immunos Building, 8A Biomedical Grove, Singapore, Singapore
| | - Chandra S Verma
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, 138671, Singapore
| | - E Birgitte Lane
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore.
- Skin Research Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Singapore, 138648, Singapore.
| |
Collapse
|
2
|
Perl AL, Pokorny JL, Green KJ. Desmosomes at a glance. J Cell Sci 2024; 137:jcs261899. [PMID: 38940346 PMCID: PMC11234380 DOI: 10.1242/jcs.261899] [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] [Indexed: 06/29/2024] Open
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
|
3
|
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
|
4
|
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] [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
|
5
|
Lu L, Ye Z, Zhang R, Olsen JV, Yuan Y, Mao Y. ETD-Based Proteomic Profiling Improves Arginine Methylation Identification and Reveals Novel PRMT5 Substrates. J Proteome Res 2024; 23:1014-1027. [PMID: 38272855 DOI: 10.1021/acs.jproteome.3c00724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
Protein arginine methylations are important post-translational modifications (PTMs) in eukaryotes, regulating many biological processes. However, traditional collision-based mass spectrometry methods inevitably cause neutral losses of methylarginines, preventing the deep mining of biologically important sites. Herein we developed an optimized mass spectrometry workflow based on electron-transfer dissociation (ETD) with supplemental activation for proteomic profiling of arginine methylation in human cells. Using symmetric dimethylarginine (sDMA) as an example, we show that the ETD-based optimized workflow significantly improved the identification and site localization of sDMA. Quantitative proteomics identified 138 novel sDMA sites as potential PRMT5 substrates in HeLa cells. Further biochemical studies on SERBP1, a newly identified PRMT5 substrate, confirmed the coexistence of sDMA and asymmetric dimethylarginine in the central RGG/RG motif, and loss of either methylation caused increased the recruitment of SERBP1 to stress granules under oxidative stress. Overall, our optimized workflow not only enabled the identification and localization of extensive, nonoverlapping sDMA sites in human cells but also revealed novel PRMT5 substrates whose sDMA may play potentially important biological functions.
Collapse
Affiliation(s)
- Lingzi Lu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Sun Yat-sen University, Guangzhou 510006, China
| | - Zilu Ye
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Rou Zhang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Sun Yat-sen University, Guangzhou 510006, China
| | - Jesper V Olsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Yanqiu Yuan
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Sun Yat-sen University, Guangzhou 510006, China
| | - Yang Mao
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
6
|
Perl AL, Koetsier JL, Green KJ. PP2A-B55alpha controls keratinocyte adhesion through dephosphorylation of the Desmoplakin C-terminus. Sci Rep 2023; 13:12720. [PMID: 37543698 PMCID: PMC10404246 DOI: 10.1038/s41598-023-37874-8] [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: 11/07/2022] [Accepted: 06/28/2023] [Indexed: 08/07/2023] Open
Abstract
Critical for the maintenance of epidermal integrity and function are attachments between intermediate filaments (IF) and intercellular junctions called desmosomes. The desmosomal cytoplasmic plaque protein desmoplakin (DP) is essential for anchoring IF to the junction. DP-IF interactions are regulated by a phospho-regulatory motif within the DP C-terminus controlling keratinocyte intercellular adhesion. Here we identify the protein phosphatase 2A (PP2A)-B55α holoenzyme as the major serine/threonine phosphatase regulating DP's C-terminus and consequent intercellular adhesion. Using a combination of chemical and genetic approaches, we show that the PP2A-B55α holoenzyme interacts with DP at intercellular membranes in 2D- and 3D- epidermal models and human skin samples. Our experiments demonstrate that PP2A-B55α regulates the phosphorylation status of junctional DP and is required for maintaining strong desmosome-mediated intercellular adhesion. These data identify PP2A-B55α as part of a regulatory module capable of tuning intercellular adhesion strength and a candidate disease target in desmosome-related disorders of the skin and heart.
Collapse
Affiliation(s)
- Abbey L Perl
- Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave., Chicago, IL, 60611, USA
| | - Jennifer L Koetsier
- Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave., Chicago, IL, 60611, USA
| | - Kathleen J Green
- Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave., Chicago, IL, 60611, USA.
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
7
|
Vielmuth F, Radeva MY, Yeruva S, Sigmund AM, Waschke J. cAMP: A master regulator of cadherin-mediated binding in endothelium, epithelium and myocardium. Acta Physiol (Oxf) 2023; 238:e14006. [PMID: 37243909 DOI: 10.1111/apha.14006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/05/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
Regulation of cadherin-mediated cell adhesion is crucial not only for maintaining tissue integrity and barrier function in the endothelium and epithelium but also for electromechanical coupling within the myocardium. Therefore, loss of cadherin-mediated adhesion causes various disorders, including vascular inflammation and desmosome-related diseases such as the autoimmune blistering skin dermatosis pemphigus and arrhythmogenic cardiomyopathy. Mechanisms regulating cadherin-mediated binding contribute to the pathogenesis of diseases and may also be used as therapeutic targets. Over the last 30 years, cyclic adenosine 3',5'-monophosphate (cAMP) has emerged as one of the master regulators of cell adhesion in endothelium and, more recently, also in epithelial cells as well as in cardiomyocytes. A broad spectrum of experimental models from vascular physiology and cell biology applied by different generations of researchers provided evidence that not only cadherins of endothelial adherens junctions (AJ) but also desmosomal contacts in keratinocytes and the cardiomyocyte intercalated discs are central targets in this scenario. The molecular mechanisms involve protein kinase A- and exchange protein directly activated by cAMP-mediated regulation of Rho family GTPases and S665 phosphorylation of the AJ and desmosome adaptor protein plakoglobin. In line with this, phosphodiesterase 4 inhibitors such as apremilast have been proposed as a therapeutic strategy to stabilize cadherin-mediated adhesion in pemphigus and may also be effective to treat other disorders where cadherin-mediated binding is compromised.
Collapse
Affiliation(s)
- Franziska Vielmuth
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Mariya Y Radeva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Sunil Yeruva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Anna M Sigmund
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| |
Collapse
|
8
|
Pantou MP, Gourzi P, Vlagkouli V, Papatheodorou E, Tsoutsinos A, Nyktari E, Degiannis D, Anastasakis A. A truncating variant altering the extreme C-terminal region of desmoplakin (DSP) suggests the crucial functional role of the region: a case report study. BMC Med Genomics 2023; 16:95. [PMID: 37143080 PMCID: PMC10158133 DOI: 10.1186/s12920-023-01527-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Homozygous truncating mutations located in the C-terminal region of the desmoplakin gene (DSP) are known to mainly cause Carvajal syndrome, an autosomal recessive syndromic form of arrhythmogenic cardiomyopathy with an extra-cardiac cutaneous phenotype. CASE PRESENTATION Here we describe a female proband with a documented arrhythmogenic left ventricular cardiomyopathy and a syncopal episode at the age of 13, who was found homozygous for the novel DSP variant: NM_004415.4:c.8586delC, p.(Ser2863Hisfs*20) at the extreme C-terminal region of the protein, just 8 amino acids upstream the stop codon. She did not have any of the typical dermatological symptoms that characterize Carvajal syndrome. Her brother had died suddenly at the age of 18 during exercise and was found homozygous for the same variant at the post-mortem, while their parents were heterozygous. The region of origin of both parents was the same geographic area of Greece, but they were not aware of any common ancestor. Detailed clinical examination revealed that the mother displayed a mild arrhythmic phenotype, while the father was asymptomatic. CONCLUSION These observations pinpoint to a significant functional role of the extreme C-terminal tail of the protein.
Collapse
Affiliation(s)
- Malena P Pantou
- Molecular Immunopathology, Histocompatibility and Genetics Laboratory, Onassis Cardiac Surgery Center, Kallithea, Greece.
| | - Polyxeni Gourzi
- Molecular Immunopathology, Histocompatibility and Genetics Laboratory, Onassis Cardiac Surgery Center, Kallithea, Greece
| | - Vasiliki Vlagkouli
- Unit of Inherited and Rare Cardiovascular Diseases, Onassis Cardiac Surgery Center, Kallithea, Greece
| | - Efstathios Papatheodorou
- Unit of Inherited and Rare Cardiovascular Diseases, Onassis Cardiac Surgery Center, Kallithea, Greece
| | - Alexandros Tsoutsinos
- Department of Pediatric Cardiology, Onassis Cardiac Surgery Center, Kallithea, Greece
| | - Eva Nyktari
- CMR Unit, Onassis Cardiac Surgery Center, Kallithea, Greece
| | - Dimitrios Degiannis
- Molecular Immunopathology, Histocompatibility and Genetics Laboratory, Onassis Cardiac Surgery Center, Kallithea, Greece
| | - Aris Anastasakis
- Unit of Inherited and Rare Cardiovascular Diseases, Onassis Cardiac Surgery Center, Kallithea, Greece
| |
Collapse
|
9
|
Yeruva S, Körber L, Hiermaier M, Egu DT, Kempf E, Waschke J. Cholinergic signaling impairs cardiomyocyte cohesion. Acta Physiol (Oxf) 2022; 236:e13881. [PMID: 36039679 DOI: 10.1111/apha.13881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 01/29/2023]
Abstract
AIM Cardiac autonomic nervous system (ANS) dysregulation is a hallmark of several cardiovascular diseases. Adrenergic signaling enhanced cardiomyocyte cohesion via PKA-mediated plakoglobin phosphorylation at serine 665, referred to as positive adhesiotropy. This study investigated cholinergic regulation of cardiomyocyte cohesion using muscarinic receptor agonist carbachol (CCH). METHODS Dissociation assays, Western blot analysis, immunostaining, atomic force microscopy (AFM), immunoprecipitation, transmission electron microscopy (TEM), triton assays, and siRNA knockdown of genes were performed in either HL-1 cells or plakoglobin (PG) wild type (Jup+/+ ) and knockout (Jup-/- ) mice, which served as a model for arrhythmogenic cardiomyopathy. RESULTS In HL-1 cells grown in norepinephrine (NE)-containing medium for baseline adrenergic stimulation, and murine cardiac slice cultures from Jup+/+ and Jup-/- mice CCH treatment impaired cardiomyocyte cohesion. Immunostainings and AFM experiments revealed that CCH reduced desmoglein 2 (DSG2) localization and binding at cell borders. Furthermore, CCH reduced intercalated disc plaque thickness in both Jup+/+ and Jup-/- mice, evidenced by TEM analysis. Immunoprecipitation experiments in HL-1 cells revealed no changes in DSG2 interaction with desmoplakin (DP), plakophilin 2 (PKP2), PG, and desmin (DES) after CCH treatment. However, knockdown of any of the above proteins abolished CCH-mediated loss of cardiomyocyte cohesion. Furthermore, in HL-1 cells, CCH inhibited adrenergic-stimulated ERK phosphorylation but not PG phosphorylation at serine 665. In addition, CCH activated the AKT/GSK-3β axis in the presence of NE. CONCLUSION Our results demonstrate that cholinergic signaling antagonizes the positive effect of adrenergic signaling on cardiomyocyte cohesion and thus causes negative adhesiotropy independent of PG phosphorylation.
Collapse
Affiliation(s)
- Sunil Yeruva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University (LMU) Munich, Munich, Germany
| | - Lars Körber
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University (LMU) Munich, Munich, Germany
| | - Matthias Hiermaier
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University (LMU) Munich, Munich, Germany
| | - Desalegn T Egu
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University (LMU) Munich, Munich, Germany
| | - Ellen Kempf
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University (LMU) Munich, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University (LMU) Munich, Munich, Germany
| |
Collapse
|
10
|
Towards a Better Understanding of Genotype-Phenotype Correlations and Therapeutic Targets for Cardiocutaneous Genes: The Importance of Functional Studies above Prediction. Int J Mol Sci 2022; 23:ijms231810765. [PMID: 36142674 PMCID: PMC9503274 DOI: 10.3390/ijms231810765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Genetic variants in gene-encoding proteins involved in cell−cell connecting structures, such as desmosomes and gap junctions, may cause a skin and/or cardiac phenotype, of which the combination is called cardiocutaneous syndrome. The cardiac phenotype is characterized by cardiomyopathy and/or arrhythmias, while the skin particularly displays phenotypes such as keratoderma, hair abnormalities and skin fragility. The reported variants associated with cardiocutaneous syndrome, in genes DSP, JUP, DSC2, KLHL24, GJA1, are classified by interpretation guidelines from the American College of Medical Genetics and Genomics. The genotype−phenotype correlation, however, remains poorly understood. By providing an overview of variants that are assessed for a functional protein pathology, we show that this number (n = 115) is low compared to the number of variants that are assessed by in silico algorithms (>5000). As expected, there is a mismatch between the prediction of variant pathogenicity and the prediction of the functional effect compared to the real functional evidence. Aiding to improve genotype−phenotype correlations, we separate variants into ‘protein reducing’ or ‘altered protein’ variants and provide general conclusions about the skin and heart phenotype involved. We conclude by stipulating that adequate prognoses can only be given, and targeted therapies can only be designed, upon full knowledge of the protein pathology through functional investigation.
Collapse
|
11
|
An X, Lee J, Kim GH, Kim HJ, Pyo HJ, Kwon I, Cho H. Modulation of I Ks channel-PIP 2 interaction by PRMT1 plays a critical role in the control of cardiac repolarization. J Cell Physiol 2022; 237:3069-3079. [PMID: 35580065 PMCID: PMC9543859 DOI: 10.1002/jcp.30775] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/18/2022] [Accepted: 04/25/2022] [Indexed: 11/12/2022]
Abstract
Recent studies have shown that protein arginine methyltransferase 1 (PRMT1) is highly expressed in the human heart, and loss of PRMT1 contributes to cardiac remodeling in the heart failure. However, the functional importance of PRMT1 in cardiac ion channels remains uncertain. The slow activating delayed rectifier K+ (IKs) channel is a cardiac K+ channel composed of KCNQ1 and KCNE1 subunits and is a new therapeutic target for treating lethal arrhythmias in many cardiac pathologies, especially heart failure. Here, we demonstrate that PRMT1 is a critical regulator of the IKs channel and cardiac rhythm. In the guinea pig ventricular myocytes, treatment with furamidine, a PRMT1‐specific inhibitor, prolonged the action potential duration (APD). We further show that this APD prolongation was attributable to IKs reduction. In HEK293T cells expressing human KCNQ1 and KCNE1, inhibiting PRMT1 via furamidine reduced IKs and concurrently decreased the arginine methylation of KCNQ1, a pore‐forming α‐subunit. Evidence presented here indicates that furamidine decreased IKs mainly by lowering the affinity of IKs channels for the membrane phospholipid, phosphatidylinositol 4,5‐bisphosphate (PIP2), which is crucial for pore opening. Finally, applying exogenous PIP2 to cardiomyocytes prevented the furamidine‐induced IKs reduction and APD prolongation. Taken together, these results indicate that PRMT1 positively regulated IKs activity through channel–PIP2 interaction, thereby restricting excessive cardiac action potential.
Collapse
Affiliation(s)
- Xue An
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Jiwon Lee
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Ga Hye Kim
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Hyun-Ji Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Hyun-Jeong Pyo
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Ilmin Kwon
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Hana Cho
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| |
Collapse
|
12
|
Büchau F, Vielmuth F, Waschke J, Magin TM. Bidirectional regulation of desmosome hyperadhesion by keratin isotypes and desmosomal components. Cell Mol Life Sci 2022; 79:223. [PMID: 35380280 PMCID: PMC8983532 DOI: 10.1007/s00018-022-04244-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/04/2022] [Accepted: 03/10/2022] [Indexed: 11/30/2022]
Abstract
Desmosomes are intercellular junctions which mediate cohesion and communication in tissues exposed to mechanical strain by tethering the intermediate filament cytoskeleton to the plasma membrane. While mature desmosomes are characterized by a hyperadhesive, Ca2+-independent state, they transiently loose this state during wound healing, pathogenesis and tissue regeneration. The mechanisms controlling the hyperadhesive state remain incompletely understood. Here, we show that upon Ca2+-induced keratinocyte differentiation, expression of keratin 17 (K17) prevents the formation of stable and hyperadhesive desmosomes, accompanied by a significant reduction of desmoplakin (DP), plakophilin-1 (PKP1), desmoglein-1 (Dsg1) and -3 (Dsg3) at intercellular cell borders. Atomic force microscopy revealed that both increased binding strength of desmoglein-3 molecules and amount of desmoglein-3 oligomers, known hallmarks of hyperadhesion, were reduced in K17- compared to K14-expressing cells. Importantly, overexpression of Dsg3 or DPII enhanced their localization at intercellular cell borders and increased the formation of Dsg3 oligomers, resulting in stable, hyperadhesive desmosomes despite the presence of K17. Notably, PKP1 was enriched in these desmosomes. Quantitative image analysis revealed that DPII overexpression contributed to desmosome hyperadhesion by increasing the abundance of K5/K17-positive keratin filaments in the proximity of desmosomes enriched in desmoglein-3. Thus, our data show that hyperadhesion can result from recruitment of keratin isotypes K5/K17 to desmosomes or from enhanced expression of DP and Dsg3 irrespective of keratin composition. The notion that hyperadhesive desmosomes failed to form in the absence of keratins underscores the essential role of keratins and suggest bidirectional control mechanisms at several levels.
Collapse
Affiliation(s)
- Fanny Büchau
- Institute of Biology, Division of Cell and Developmental Biology, University of Leipzig, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany.
| | - Franziska Vielmuth
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Thomas M Magin
- Institute of Biology, Division of Cell and Developmental Biology, University of Leipzig, Philipp-Rosenthal-Straße 55, 04103, Leipzig, Germany
| |
Collapse
|
13
|
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: 34] [Impact Index Per Article: 17.0] [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
|
14
|
Beggs RR, Rao TC, Dean WF, Kowalczyk AP, Mattheyses AL. Desmosomes undergo dynamic architectural changes during assembly and maturation. Tissue Barriers 2022; 10:2017225. [PMID: 34983311 PMCID: PMC9621066 DOI: 10.1080/21688370.2021.2017225] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Desmosomes are macromolecular cell-cell junctions critical for maintaining adhesion and resisting mechanical stress in epithelial tissue. Desmosome assembly and the relationship between maturity and molecular architecture are not well understood. To address this, we employed a calcium switch assay to synchronize assembly followed by quantification of desmosome nanoscale organization using direct Stochastic Optical Reconstruction Microscopy (dSTORM). We found that the organization of the desmoplakin rod/C-terminal junction changed over the course of maturation, as indicated by a decrease in the plaque-to-plaque distance, while the plaque length increased. In contrast, the desmoplakin N-terminal domain and plakoglobin organization (plaque-to-plaque distance) were constant throughout maturation. This structural rearrangement of desmoplakin was concurrent with desmosome maturation measured by E-cadherin exclusion and increased adhesive strength. Using two-color dSTORM, we showed that while the number of individual E-cadherin containing junctions went down with the increasing time in high Ca2+, they maintained a wider desmoplakin rod/C-terminal plaque-to-plaque distance. This indicates that the maturation state of individual desmosomes can be identified by their architectural organization. We confirmed these architectural changes in another model of desmosome assembly, cell migration. Desmosomes in migrating cells, closest to the scratch where they are assembling, were shorter, E-cadherin enriched, and had wider desmoplakin rod/C-terminal plaque-to-plaque distances compared to desmosomes away from the wound edge. Key results were demonstrated in three cell lines representing simple, transitional, and stratified epithelia. Together, these data suggest that there is a set of architectural programs for desmosome maturation, and we hypothesize that desmoplakin architecture may be a contributing mechanism to regulating adhesive strength.
Collapse
Affiliation(s)
- Reena R Beggs
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tejeshwar C Rao
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - William F Dean
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Andrew P Kowalczyk
- Departments of Dermatology and Cellular and Molecular Physiology, Pennsylvania State College of Medicine, Hershey, PA, USA
| | - Alexa L Mattheyses
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
15
|
Mohammed F, Odintsova E, Chidgey M. Missense Mutations in Desmoplakin Plakin Repeat Domains Have Dramatic Effects on Domain Structure and Function. Int J Mol Sci 2022; 23:ijms23010529. [PMID: 35008956 PMCID: PMC8745463 DOI: 10.3390/ijms23010529] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/24/2021] [Accepted: 12/29/2021] [Indexed: 02/06/2023] Open
Abstract
Plakin repeat domains (PRDs) are globular modules that mediate the interaction of plakin proteins with the intermediate filament (IF) cytoskeleton. These associations are vital for maintaining tissue integrity in cardiac muscle and epithelial tissues. PRDs are subject to mutations that give rise to cardiomyopathies such as arrhythmogenic right ventricular cardiomyopathy, characterised by ventricular arrhythmias and associated with an increased risk of sudden heart failure, and skin blistering diseases. Herein, we have examined the functional and structural effects of 12 disease-linked missense mutations, identified from the human gene mutation database, on the PRDs of the desmosomal protein desmoplakin. Five mutations (G2056R and E2193K in PRD-A, G2338R and G2375R in PRD-B and G2647D in PRD-C) rendered their respective PRD proteins either fully or partially insoluble following expression in bacterial cells. Each of the residues affected are conserved across plakin family members, inferring a crucial role in maintaining the structural integrity of the PRD. In transfected HeLa cells, the mutation G2375R adversely affected the targeting of a desmoplakin C-terminal construct containing all three PRDs to vimentin IFs. The deletion of PRD-B and PRD-C from the construct compromised its targeting to vimentin. Bioinformatic and structural modelling approaches provided multiple mechanisms by which the disease-causing mutations could potentially destabilise PRD structure and compromise cytoskeletal linkages. Overall, our data highlight potential molecular mechanisms underlying pathogenic missense mutations and could pave the way for informing novel curative interventions targeting cardiomyopathies and skin blistering disorders.
Collapse
Affiliation(s)
- Fiyaz Mohammed
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK;
| | - Elena Odintsova
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - Martyn Chidgey
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK;
- Correspondence:
| |
Collapse
|
16
|
Proteomic characterization of GSK3β knockout shows altered cell adhesion and metabolic pathway utilisation in colorectal cancer cells. PLoS One 2021; 16:e0246707. [PMID: 34739494 PMCID: PMC8570494 DOI: 10.1371/journal.pone.0246707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 10/01/2021] [Indexed: 11/22/2022] Open
Abstract
Glycogen-specific kinase (GSK3β) is an integral regulator of the Wnt signalling pathway as well as many other diverse signalling pathways and processes. Dys-regulation of GSK3β is implicated in many different pathologies, including neurodegenerative disorders as well as many different tumour types. In the context of tumour development, GSK3β has been shown to play both oncogenic and tumour suppressor roles, depending upon tissue, signalling environment or disease progression. Although multiple substrates of the GSK3β kinase have been identified, the wider protein networks within which GSK3β participates are not well known, and the consequences of these interactions not well understood. In this study, LC-MS/MS expression analysis was performed using knockout GSK3β colorectal cancer cells and isogenic controls in colorectal cancer cell lines carrying dominant stabilizing mutations of β-catenin. Consistent with the role of GSK3β, we found that β-catenin levels and canonical Wnt activity are unaffected by knockout of GSK3β and therefore used this knockout cell model to identify other processes in which GSK3β is implicated. Quantitative proteomic analysis revealed perturbation of proteins involved in cell-cell adhesion, and we characterized the phenotype and altered proteomic profiles associated with this. We also characterized the perturbation of metabolic pathways resulting from GSK3β knockout and identified defects in glycogen metabolism. In summary, using a precision colorectal cancer cell-line knockout model with constitutively activated β-catenin we identified several of the diverse pathways and processes associated with GSK3β function.
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: 12] [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
|
Müller L, Hatzfeld M, Keil R. Desmosomes as Signaling Hubs in the Regulation of Cell Behavior. Front Cell Dev Biol 2021; 9:745670. [PMID: 34631720 PMCID: PMC8495202 DOI: 10.3389/fcell.2021.745670] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [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
|
19
|
Mohammed F, Chidgey M. Desmosomal protein structure and function and the impact of disease-causing mutations. J Struct Biol 2021; 213:107749. [PMID: 34033898 DOI: 10.1016/j.jsb.2021.107749] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 11/26/2022]
Abstract
In this graphical review we focus on the structural characteristics of desmosomal proteins, their interactions with each other and with the intermediate filament cytoskeleton. The wealth of structural information that is now available allows predictions to be made about the pathogenic effect of disease-causing mutations. We have selected representative examples of missense mutations that are buried, semi-buried or surface exposed, and demonstrate how such variants could affect the structural fold of desmosomal proteins that are expressed in the heart. We explain how such alterations could compromise desmosomal adhesion, resulting in life threatening diseases including arrhythmogenic right ventricular cardiomyopathy.
Collapse
Affiliation(s)
- Fiyaz Mohammed
- Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK.
| | - Martyn Chidgey
- Institute of Clinical Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
20
|
Raya-Sandino A, Luissint AC, Kusters DHM, Narayanan V, Flemming S, Garcia-Hernandez V, Godsel LM, Green KJ, Hagen SJ, Conway DE, Parkos CA, Nusrat A. Regulation of intestinal epithelial intercellular adhesion and barrier function by desmosomal cadherin desmocollin-2. Mol Biol Cell 2021; 32:753-768. [PMID: 33596089 PMCID: PMC8108520 DOI: 10.1091/mbc.e20-12-0775] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/26/2022] Open
Abstract
The role of desmosomal cadherin desmocollin-2 (Dsc2) in regulating barrier function in intestinal epithelial cells (IECs) is not well understood. Here, we report the consequences of silencing Dsc2 on IEC barrier function in vivo using mice with inducible intestinal-epithelial-specific Dsc2 knockdown (KD) (Dsc2ERΔIEC). While the small intestinal gross architecture was maintained, loss of epithelial Dsc2 influenced desmosomal plaque structure, which was smaller in size and had increased intermembrane space between adjacent epithelial cells. Functional analysis revealed that loss of Dsc2 increased intestinal permeability in vivo, supporting a role for Dsc2 in the regulation of intestinal epithelial barrier function. These results were corroborated in model human IECs in which Dsc2 KD resulted in decreased cell-cell adhesion and impaired barrier function. It is noteworthy that Dsc2 KD cells exhibited delayed recruitment of desmoglein-2 (Dsg2) to the plasma membrane after calcium switch-induced intercellular junction reassembly, while E-cadherin accumulation was unaffected. Mechanistically, loss of Dsc2 increased desmoplakin (DP I/II) protein expression and promoted intermediate filament interaction with DP I/II and was associated with enhanced tension on desmosomes as measured by a Dsg2-tension sensor. In conclusion, we provide new insights on Dsc2 regulation of mechanical tension, adhesion, and barrier function in IECs.
Collapse
Affiliation(s)
- Arturo Raya-Sandino
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Anny-Claude Luissint
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Dennis H. M. Kusters
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Vani Narayanan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284
| | - Sven Flemming
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | | | - Lisa M. Godsel
- Departments of Pathology and Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Kathleen J. Green
- Departments of Pathology and Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611
| | - Susan J. Hagen
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA 02115
| | - Daniel E. Conway
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284
| | - Charles A. Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Asma Nusrat
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
21
|
Hao Y, Bates S, Mou H, Yun JH, Pham B, Liu J, Qiu W, Guo F, Morrow JD, Hersh CP, Benway CJ, Gong L, Zhang Y, Rosas IO, Cho MH, Park JA, Castaldi PJ, Du F, Zhou X. Genome-Wide Association Study: Functional Variant rs2076295 Regulates Desmoplakin Expression in Airway Epithelial Cells. Am J Respir Crit Care Med 2020; 202:1225-1236. [PMID: 32551799 PMCID: PMC7605184 DOI: 10.1164/rccm.201910-1958oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 06/18/2020] [Indexed: 12/15/2022] Open
Abstract
Rationale: Genetic association studies have identified rs2076295 in association with idiopathic pulmonary fibrosis (IPF). We hypothesized that rs2076295 is the functional variant regulating DSP (desmoplakin) expression in human bronchial epithelial cells, and DSP regulates extracellular matrix-related gene expression and cell migration, which is relevant to IPF development.Objectives: To determine whether rs2076295 regulates DSP expression and the function of DSP in airway epithelial cells.Methods: Using CRISPR (clustered regularly interspaced short palindromic repeat)/Cas9 editing (including regional deletion, indel, CRISPR interference, and single-base editing), we modified rs2076295 and measured DSP expression in edited 16HBE14o- and primary airway epithelial cells. Cellular integrity, migration, and genome-wide gene expression changes were examined in 16HBE14o- single colonies with DSP knockout. The expression of DSP and its relevant matrix genes was measured by quantitative PCR and also analyzed in single-cell RNA-sequencing data from control and IPF lungs.Measurements and Main Results:DSP is expressed predominantly in bronchial and alveolar epithelial cells, with reduced expression in alveolar epithelial cells in IPF lungs. The deletion of the DNA region-spanning rs2076295 led to reduced expression of DSP, and the edited rs2076295GG 16HBE14o- line has lower expression of DSP than the rs2076295TT lines. Knockout of DSP in 16HBE14o- cells decreased transepithelial resistance but increased cell migration, with increased expression of extracellular matrix-related genes, including MMP7 and MMP9. Silencing of MMP7 and MMP9 abolished increased migration in DSP-knockout cells.Conclusions: rs2076295 regulates DSP expression in human airway epithelial cells. The loss of DSP enhances extracellular matrix-related gene expression and promotes cell migration, which may contribute to the pathogenesis of IPF.
Collapse
Affiliation(s)
- Yuan Hao
- Channing Division of Network Medicine and
| | | | - Hongmei Mou
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts; and
| | | | - Betty Pham
- Channing Division of Network Medicine and
| | | | | | - Feng Guo
- Channing Division of Network Medicine and
| | | | | | | | - Lu Gong
- Channing Division of Network Medicine and
| | - Yihan Zhang
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts; and
| | - Ivan O. Rosas
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Jin-Ah Park
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | - Fei Du
- Channing Division of Network Medicine and
| | | |
Collapse
|
22
|
He L, Hu Z, Sun Y, Zhang M, Zhu H, Jiang L, Zhang Q, Mu D, Zhang J, Gu L, Yang Y, Pan FY, Jia S, Guo Z. PRMT1 is critical to FEN1 expression and drug resistance in lung cancer cells. DNA Repair (Amst) 2020; 95:102953. [PMID: 32861926 DOI: 10.1016/j.dnarep.2020.102953] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/30/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022]
Abstract
The up-regulation of PRMT1 is critical to the cell growth and cancer progression of lung cancer cells. In our research, we found that PRMT1 is important to the DNA repair ability and drug resistance of lung cancer cells. To demonstrate the functions of PRMT1, we identified Flap endonuclease 1 (FEN1) as a post-translationally modified downstream target protein of PRMT1. As a major component of Base Excision Repair pathway, FEN1 plays an important role in DNA replication and DNA damage repair. However, the detailed mechanism of FEN1 up-regulation in lung cancer cells remains unclear. In our study, we identified PRMT1 as a key factor that maintains the high expression levels of FEN1, which is critical to the DNA repair ability and the chemotherapeutic drug resistance of lung cancer cells.
Collapse
Affiliation(s)
- Lingfeng He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Zhigang Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Yuling Sun
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Miaomiao Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Hongqiao Zhu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Longwei Jiang
- Jinlin Hospital of Nanjing University, Nanjing, 210002, China
| | - Qi Zhang
- Department of Infectious Diseases, Nanjing Liuhe District People's Hospital Affiliated to Yangzhou University, Nanjing, 210012, China
| | - Dan Mu
- Affiliated Drum Tower Hospital, Nanjing University School of Medicine, 210008, Nanjing, China
| | - Jing Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Lili Gu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Yang Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China
| | - Fei-Yan Pan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China.
| | - Shaochang Jia
- Jinlin Hospital of Nanjing University, Nanjing, 210002, China.
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 WenYuan Road, Nanjing, 210023, China.
| |
Collapse
|
23
|
Mohammed F, Trieber C, Overduin M, Chidgey M. Molecular mechanism of intermediate filament recognition by plakin proteins. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118801. [PMID: 32712070 DOI: 10.1016/j.bbamcr.2020.118801] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/15/2020] [Accepted: 07/20/2020] [Indexed: 12/26/2022]
Abstract
The plakin family of cytolinkers interacts with intermediate filaments (IFs) through plakin repeat domain (PRD) and linker modules. Recent structure/function studies have established the molecular basis of envoplakin-PRD and periplakin-linker interactions with vimentin. Both plakin modules share a broad basic groove which recognizes acidic rod elements on IFs, a mechanism that is applicable to other plakin family members. This review postulates a universal IF engagement mechanism that illuminates the specific effects of pathogenic mutations associated with diseases including arrhythmogenic right ventricular cardiomyopathy, and reveals how diverse plakin proteins offer tailored IF tethering to ensure stable, dynamic and regulated cellular structures.
Collapse
Affiliation(s)
- Fiyaz Mohammed
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK.
| | - Catharine Trieber
- Department of Biochemistry, Faculty of Medicine and Dentistry, 474 Medical Sciences Building, University of Alberta, Edmonton, Alberta T6G 2H7, Canada.
| | - Michael Overduin
- Department of Biochemistry, Faculty of Medicine and Dentistry, 474 Medical Sciences Building, University of Alberta, Edmonton, Alberta T6G 2H7, Canada.
| | - Martyn Chidgey
- Institute of Clinical Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
24
|
Bartle EI, Rao TC, Beggs RR, Dean WF, Urner TM, Kowalczyk AP, Mattheyses AL. Protein exchange is reduced in calcium-independent epithelial junctions. J Cell Biol 2020; 219:151763. [PMID: 32399559 PMCID: PMC7265307 DOI: 10.1083/jcb.201906153] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 01/29/2020] [Accepted: 03/11/2020] [Indexed: 12/29/2022] Open
Abstract
Desmosomes are cell–cell junctions that provide mechanical integrity to epithelial and cardiac tissues. Desmosomes have two distinct adhesive states, calcium-dependent and hyperadhesive, which balance tissue plasticity and strength. A highly ordered array of cadherins in the adhesive interface is hypothesized to drive hyperadhesion, but how desmosome structure confers adhesive state is still elusive. We employed fluorescence polarization microscopy to show that cadherin order is not required for hyperadhesion induced by pharmacologic and genetic approaches. FRAP experiments in cells treated with the PKCα inhibitor Gö6976 revealed that cadherins, plakoglobin, and desmoplakin have significantly reduced exchange in and out of hyperadhesive desmosomes. To test whether this was a result of enhanced keratin association, we used the desmoplakin mutant S2849G, which conferred reduced protein exchange. We propose that inside-out regulation of protein exchange modulates adhesive function, whereby proteins are “locked in” to hyperadhesive desmosomes while protein exchange confers plasticity on calcium-dependent desmosomes, thereby providing rapid control of adhesion.
Collapse
Affiliation(s)
- Emily I Bartle
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Tejeshwar C Rao
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Reena R Beggs
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - William F Dean
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Tara M Urner
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Andrew P Kowalczyk
- Departments of Cell Biology and Dermatology, Emory University, Atlanta, GA
| | - Alexa L Mattheyses
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
25
|
Abstract
Cadherin-based cell-cell junctions help metazoans form polarized sheets of cells, which are necessary for the development of organs and the compartmentalization of functions. The components of the protein complexes that generate cadherin-based junctions have ancient origins, with conserved elements shared between animals as diverse as sponges and vertebrates. In invertebrates, the formation and function of epithelial sheets depends on classical cadherin-containing adherens junctions, which link actin to the plasma membrane through α-, β- and p120 catenins. Vertebrates also have a new type of cadherin-based intercellular junction called the desmosome, which allowed for the creation of more complex and effective tissue barriers against environmental stress. While desmosomes have a molecular blueprint that is similar to that of adherens junctions, desmosomal cadherins - called desmogleins and desmocollins - link intermediate filaments (IFs) rather than actin to the plasma membrane through protein complexes comprising relatives of β-catenin (plakoglobin) and p120 catenin (plakophilins). In turn, desmosomal catenins interact with members of the IF-binding plakin family to create the desmosome-IF linking complex. In this Minireview, we discuss when and how desmosomal components evolved, and how their ability to anchor the highly elastic and tough IF cytoskeleton endowed vertebrates with robust tissues capable of not only resisting but also properly responding to environmental stress.
Collapse
Affiliation(s)
- Kathleen J Green
- Departments of Pathology and Dermatology, Feinberg School of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| | - Quinn Roth-Carter
- Departments of Pathology and Dermatology, Feinberg School of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA
| | - Carien M Niessen
- Department of Dermatology, Cologne Excellence Cluster on Stress Responses in Aging-associated Diseases (CECAD), Center for Molecule Medicine Cologne, University of Cologne, Cologne, Germany
| | - Scott A Nichols
- Department of Biological Sciences, 2101 E. Wesley Ave. SGM 203, University of Denver, CO 80210, USA.
| |
Collapse
|
26
|
Poller W, Haas J, Klingel K, Kühnisch J, Gast M, Kaya Z, Escher F, Kayvanpour E, Degener F, Opgen-Rhein B, Berger F, Mochmann HC, Skurk C, Heidecker B, Schultheiss HP, Monserrat L, Meder B, Landmesser U, Klaassen S. Familial Recurrent Myocarditis Triggered by Exercise in Patients With a Truncating Variant of the Desmoplakin Gene. J Am Heart Assoc 2020; 9:e015289. [PMID: 32410525 PMCID: PMC7660888 DOI: 10.1161/jaha.119.015289] [Citation(s) in RCA: 40] [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] [Indexed: 12/19/2022]
Abstract
Background Variants of the desmosomal protein desmoplakin are associated with arrhythmogenic cardiomyopathy, an important cause of ventricular arrhythmias in children and young adults. Disease penetrance of desmoplakin variants is incomplete and variant carriers may display noncardiac, dermatologic phenotypes. We describe a novel cardiac phenotype associated with a truncating desmoplakin variant, likely causing mechanical instability of myocardial desmosomes. Methods and Results In 2 young brothers with recurrent myocarditis triggered by physical exercise, screening of 218 cardiomyopathy‐related genes identified the heterozygous truncating variant p.Arg1458Ter in desmoplakin. Screening for infections yielded no evidence of viral or nonviral infections. Myosin and troponin I autoantibodies were detected at high titers. Immunohistology failed to detect any residual DSP protein in endomyocardial biopsies, and none of the histologic criteria of arrhythmogenic cardiomyopathy were fulfilled. Cardiac magnetic resonance imaging revealed no features associated with right ventricular arrhythmogenic cardiomyopathy, but multifocal subepicardial late gadolinium enhancement was present in the left ventricles of both brothers. Screening of adult cardiomyopathy cohorts for truncating variants identified the rare genetic variants p.Gln307Ter, p.Tyr1391Ter, and p.Tyr1512Ter, suggesting that over subsequent decades critical genetic/exogenous modifiers drive pathogenesis from desmoplakin truncations toward different end points. Conclusions The described novel phenotype of familial recurrent myocarditis associated with a desmoplakin truncation in adolescents likely represents a serendipitously revealed subtype of arrhythmogenic cardiomyopathy. It may be caused by a distinctive adverse effect of the variant desmoplakin upon the mechanical stability of myocardial desmosomes. Variant screening is advisable to allow early detection of patients with similar phenotypes.
Collapse
Affiliation(s)
- Wolfgang Poller
- Department of Cardiology Campus Benjamin Franklin Universitätsmedizin Berlin Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT) Universitätsmedizin Berlin Germany.,German Center for Cardiovascular Research (DZHK) partner site Berlin Germany
| | - Jan Haas
- German Center for Cardiovascular Research (DZHK) partner site Heidelberg Germany.,Department of Cardiology University Hospital Heidelberg Mannheim Germany
| | - Karin Klingel
- Institute for Pathology and Neuropathology Department of Pathology University Hospital Tübingen Germany
| | - Jirko Kühnisch
- German Center for Cardiovascular Research (DZHK) partner site Berlin Germany.,Experimental and Clinical Research Center (ECRC) Universitätsmedizin Berlin Germany
| | - Martina Gast
- Department of Cardiology Campus Benjamin Franklin Universitätsmedizin Berlin Germany
| | - Ziya Kaya
- German Center for Cardiovascular Research (DZHK) partner site Heidelberg Germany.,Department of Cardiology University Hospital Heidelberg Mannheim Germany
| | - Felicitas Escher
- Department of Cardiology Campus Virchow Klinikum Universitätsmedizin Berlin Germany.,Institute for Clinical Diagnostics and Therapy (IKDT) Berlin Germany
| | - Elham Kayvanpour
- German Center for Cardiovascular Research (DZHK) partner site Heidelberg Germany.,Department of Cardiology University Hospital Heidelberg Mannheim Germany
| | - Franziska Degener
- German Center for Cardiovascular Research (DZHK) partner site Berlin Germany.,German Heart Center (DHZB) Berlin Germany
| | - Bernd Opgen-Rhein
- Department of Pediatric Cardiology Universitätsmedizin Berlin Germany
| | - Felix Berger
- German Center for Cardiovascular Research (DZHK) partner site Berlin Germany.,German Heart Center (DHZB) Berlin Germany.,Department of Pediatric Cardiology Universitätsmedizin Berlin Germany
| | | | - Carsten Skurk
- Department of Cardiology Campus Benjamin Franklin Universitätsmedizin Berlin Germany
| | - Bettina Heidecker
- Department of Cardiology Campus Benjamin Franklin Universitätsmedizin Berlin Germany
| | | | | | - Benjamin Meder
- German Center for Cardiovascular Research (DZHK) partner site Heidelberg Germany.,Department of Cardiology University Hospital Heidelberg Mannheim Germany.,Department of Genetics Stanford University School of Medicine Palo Alto CA
| | - Ulf Landmesser
- Department of Cardiology Campus Benjamin Franklin Universitätsmedizin Berlin Germany.,German Center for Cardiovascular Research (DZHK) partner site Berlin Germany.,Berlin Institute of Health Berlin Germany
| | - Sabine Klaassen
- German Center for Cardiovascular Research (DZHK) partner site Berlin Germany.,Experimental and Clinical Research Center (ECRC) Universitätsmedizin Berlin Germany.,Department of Pediatric Cardiology Universitätsmedizin Berlin Germany
| |
Collapse
|
27
|
Camors EM, Purevjav E, Jefferies JL, Saffitz JE, Gong N, Ryan TD, Lucky AW, Taylor MD, Sullivan LM, Mestroni L, Towbin JA. Early Lethality Due to a Novel Desmoplakin Variant Causing Infantile Epidermolysis Bullosa Simplex With Fragile Skin, Aplasia Cutis Congenita, and Arrhythmogenic Cardiomyopathy. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2020; 13:e002800. [PMID: 32164419 DOI: 10.1161/circgen.119.002800] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Emmanuel M Camors
- Department of Pediatrics, The Heart Institute (E.M.C., E.P., J.L.J., N.G., T.D.R., M.D.T., J.A.T.), Cincinnati Children's Hospital Medical Center, OH.,The Division of Cardiology, Department of Pediatrics, University of Tennessee Health Science Center and the Heart Institute, Le Bonheur Children's Hospital Research Center, Memphis, TN (E.M.C., E.P., J.A.T.)
| | - Enkhsaikhan Purevjav
- Department of Pediatrics, The Heart Institute (E.M.C., E.P., J.L.J., N.G., T.D.R., M.D.T., J.A.T.), Cincinnati Children's Hospital Medical Center, OH.,The Division of Cardiology, Department of Pediatrics, University of Tennessee Health Science Center and the Heart Institute, Le Bonheur Children's Hospital Research Center, Memphis, TN (E.M.C., E.P., J.A.T.)
| | - John L Jefferies
- Department of Pediatrics, The Heart Institute (E.M.C., E.P., J.L.J., N.G., T.D.R., M.D.T., J.A.T.), Cincinnati Children's Hospital Medical Center, OH
| | - Jeffrey E Saffitz
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA (J.E.S.)
| | - Nan Gong
- Department of Pediatrics, The Heart Institute (E.M.C., E.P., J.L.J., N.G., T.D.R., M.D.T., J.A.T.), Cincinnati Children's Hospital Medical Center, OH
| | - Thomas D Ryan
- Department of Pediatrics, The Heart Institute (E.M.C., E.P., J.L.J., N.G., T.D.R., M.D.T., J.A.T.), Cincinnati Children's Hospital Medical Center, OH
| | - Anne W Lucky
- The Division of Dermatology, Department of Pediatrics (A.W.L.), Cincinnati Children's Hospital Medical Center, OH
| | - Michael D Taylor
- Department of Pediatrics, The Heart Institute (E.M.C., E.P., J.L.J., N.G., T.D.R., M.D.T., J.A.T.), Cincinnati Children's Hospital Medical Center, OH
| | - Lisa M Sullivan
- Department of Pathology, University of Mississippi Medical Center, Jackson (L.M.S.)
| | - Luisa Mestroni
- The Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver Anschutz Medical Campus, Aurora (L.M.)
| | - Jeffrey A Towbin
- Department of Pediatrics, The Heart Institute (E.M.C., E.P., J.L.J., N.G., T.D.R., M.D.T., J.A.T.), Cincinnati Children's Hospital Medical Center, OH.,The Division of Cardiology, Department of Pediatrics, University of Tennessee Health Science Center and the Heart Institute, Le Bonheur Children's Hospital Research Center, Memphis, TN (E.M.C., E.P., J.A.T.)
| |
Collapse
|
28
|
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: 7.5] [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
|
29
|
Al-Hamashi AA, Diaz K, Huang R. Non-Histone Arginine Methylation by Protein Arginine Methyltransferases. Curr Protein Pept Sci 2020; 21:699-712. [PMID: 32379587 PMCID: PMC7529871 DOI: 10.2174/1389203721666200507091952] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/17/2019] [Accepted: 09/27/2019] [Indexed: 12/23/2022]
Abstract
Protein arginine methyltransferase (PRMT) enzymes play a crucial role in RNA splicing, DNA damage repair, cell signaling, and differentiation. Arginine methylation is a prominent posttransitional modification of histones and various non-histone proteins that can either activate or repress gene expression. The aberrant expression of PRMTs has been linked to multiple abnormalities, notably cancer. Herein, we review a number of non-histone protein substrates for all nine members of human PRMTs and how PRMT-mediated non-histone arginine methylation modulates various diseases. Additionally, we highlight the most recent clinical studies for several PRMT inhibitors.
Collapse
Affiliation(s)
- Ayad A. Al-Hamashi
- Department of Medicinal Chemistry and Molecular Pharmacology, Center for Cancer Research, Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, United States
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Baghdad, Bab-almoadham, Baghdad, Iraq
| | - Krystal Diaz
- Department of Medicinal Chemistry and Molecular Pharmacology, Center for Cancer Research, Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, United States
| | - Rong Huang
- Department of Medicinal Chemistry and Molecular Pharmacology, Center for Cancer Research, Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, United States
| |
Collapse
|
30
|
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: 53] [Impact Index Per Article: 10.6] [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
|
31
|
Tejeda-Muñoz N, Albrecht LV, Bui MH, De Robertis EM. Wnt canonical pathway activates macropinocytosis and lysosomal degradation of extracellular proteins. Proc Natl Acad Sci U S A 2019; 116:10402-10411. [PMID: 31061124 PMCID: PMC6534993 DOI: 10.1073/pnas.1903506116] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Canonical Wnt signaling is emerging as a major regulator of endocytosis. Wnt treatment markedly increased the endocytosis and degradation in lysosomes of BSA. In this study, we report that in addition to receptor-mediated endocytosis, Wnt also triggers the intake of large amounts of extracellular fluid by macropinocytosis, a nonreceptor-mediated actin-driven process. Macropinocytosis induction is rapid and independent of protein synthesis. In the presence of Wnt, large amounts of nutrient-rich packages such as proteins and glycoproteins were channeled into lysosomes after fusing with smaller receptor-mediated vesicles containing glycogen synthase kinase 3 (GSK3) and protein arginine ethyltransferase 1 (PRMT1), an enzyme required for canonical Wnt signaling. Addition of Wnt3a, as well as overexpression of Disheveled (Dvl), Frizzled (Fz8), or dominant-negative Axin induced endocytosis. Depletion of the tumor suppressors adenomatous polyposis coli (APC) or Axin dramatically increased macropinocytosis, defined by incorporation of the high molecular weight marker tetramethylrhodamine (TMR)-dextran and its blockage by the Na+/H+ exchanger ethylisopropyl amiloride (EIPA). Macropinocytosis was blocked by dominant-negative vacuolar protein sorting 4 (Vps4), indicating that the Wnt pathway is dependent on multivesicular body formation, a process called microautophagy. SW480 colorectal cancer cells displayed constitutive macropinocytosis and increased extracellular protein degradation in lysosomes, which were suppressed by restoring full-length APC. Accumulation of the transcriptional activator β-catenin in the nucleus of SW480 cells was inhibited by methyltransferase inhibition, EIPA, or the diuretic amiloride. The results indicate that Wnt signaling switches metabolism toward nutrient acquisition by engulfment of extracellular fluids and suggest possible treatments for Wnt-driven cancer progression.
Collapse
Affiliation(s)
- Nydia Tejeda-Muñoz
- Howard Hughes Medical Institute, University of California, Los Angeles, CA 90095-1662
- Department of Biological Chemistry, University of California, Los Angeles, CA 90095-1662
| | - Lauren V Albrecht
- Howard Hughes Medical Institute, University of California, Los Angeles, CA 90095-1662
- Department of Biological Chemistry, University of California, Los Angeles, CA 90095-1662
| | - Maggie H Bui
- Howard Hughes Medical Institute, University of California, Los Angeles, CA 90095-1662
- Department of Biological Chemistry, University of California, Los Angeles, CA 90095-1662
| | - Edward M De Robertis
- Howard Hughes Medical Institute, University of California, Los Angeles, CA 90095-1662;
- Department of Biological Chemistry, University of California, Los Angeles, CA 90095-1662
| |
Collapse
|
32
|
The Chinese Medicinal Formulation Guzhi Zengsheng Zhitongwan Modulates Chondrocyte Structure, Dynamics, and Metabolism by Controlling Multiple Functional Proteins. BIOMED RESEARCH INTERNATIONAL 2019; 2018:9847286. [PMID: 30596102 PMCID: PMC6282133 DOI: 10.1155/2018/9847286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 11/08/2018] [Indexed: 12/15/2022]
Abstract
Traditional Chinese medicine is one of the oldest medical systems in the world and has its unique principles and theories in the prevention and treatment of human diseases, which are achieved through the interactions of different types of materia medica in the form of Chinese medicinal formulations. GZZSZTW, a classical and effective Chinese medicinal formulation, was designed and created by professor Bailing Liu who is the only national medical master professor in the clinical research field of traditional Chinese medicine and skeletal diseases. GZZSZTW has been widely used in clinical settings for several decades for the treatment of joint diseases. However, the underlying molecular mechanisms are still largely unknown. In the present study, we performed quantitative proteomic analysis to investigate the effects of GZZSZTW on mouse primary chondrocytes using state-of-the-art iTRAQ technology. We demonstrated that the Chinese medicinal formulation GZZSZTW modulates chondrocyte structure, dynamics, and metabolism by controlling multiple functional proteins that are involved in the cellular processes of DNA replication and transcription, protein synthesis and degradation, cytoskeleton dynamics, and signal transduction. Thus, this study has expanded the current knowledge of the molecular mechanism of GZZSZTW treatment on chondrocytes. It has also shed new light on possible strategies to further prevent and treat cartilage-related diseases using traditional Chinese medicinal formulations.
Collapse
|
33
|
Cardiac specific PRMT1 ablation causes heart failure through CaMKII dysregulation. Nat Commun 2018; 9:5107. [PMID: 30504773 PMCID: PMC6269446 DOI: 10.1038/s41467-018-07606-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 11/02/2018] [Indexed: 02/07/2023] Open
Abstract
Dysregulation of Ca2+/calmodulin-dependent protein kinase (CaMK)II is closely linked with myocardial hypertrophy and heart failure. However, the mechanisms that regulate CaMKII activity are incompletely understood. Here we show that protein arginine methyltransferase 1 (PRMT1) is essential for preventing cardiac CaMKII hyperactivation. Mice null for cardiac PRMT1 exhibit a rapid progression to dilated cardiomyopathy and heart failure within 2 months, accompanied by cardiomyocyte hypertrophy and fibrosis. Consistently, PRMT1 is downregulated in heart failure patients. PRMT1 depletion in isolated cardiomyocytes evokes hypertrophic responses with elevated remodeling gene expression, while PRMT1 overexpression protects against pathological responses to neurohormones. The level of active CaMKII is significantly elevated in PRMT1-deficient hearts or cardiomyocytes. PRMT1 interacts with and methylates CaMKII at arginine residues 9 and 275, leading to its inhibition. Accordingly, pharmacological inhibition of CaMKII restores contractile function in PRMT1-deficient mice. Thus, our data suggest that PRMT1 is a critical regulator of CaMKII to maintain cardiac function. The mechanisms that regulate the activity of Ca2 +/calmodulin-dependent protein kinase II (CaMKII) in the context of heart failure are incompletely understood. Here the authors show that protein arginine methyltransferase 1 (PRMT1) prevents cardiac hyperactivation of CaMKII and heart failure development by methylating CaMKII at arginine residues 9 and 275.
Collapse
|
34
|
Favre B, Begré N, Bouameur JE, Lingasamy P, Conover GM, Fontao L, Borradori L. Desmoplakin interacts with the coil 1 of different types of intermediate filament proteins and displays high affinity for assembled intermediate filaments. PLoS One 2018; 13:e0205038. [PMID: 30286183 PMCID: PMC6171917 DOI: 10.1371/journal.pone.0205038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/18/2018] [Indexed: 12/04/2022] Open
Abstract
The interaction of intermediate filaments (IFs) with the cell-cell adhesion complexes desmosomes is crucial for cytoskeletal organization and cell resilience in the epidermis and heart. The intracellular desmosomal protein desmoplakin anchors IFs to the cell adhesion complexes predominantly via its four last carboxy-terminal domains (C-terminus). However, it remains unclear why the C-terminus of desmoplakin interacts with different IF types or if there are different binding affinities for each type of IFs that may influence the stability of cell-specific adhesion complexes. By yeast three-hybrid and fluorescence binding assays, we found that the coiled-coil 1 of the conserved central rod domain of the heterodimeric cytokeratins (Ks) 5 and 14 (K5/K14) was required for their interaction with the C-terminus of desmoplakin, while their unique amino head- and C-tail domains were dispensable. Similar findings were obtained in vitro with K1/K10, and the type III IF proteins desmin and vimentin. Binding assays testing the C-terminus of desmoplakin with assembled K5/K14 and desmin IFs yielded an apparent affinity in the nM range. Our findings reveal that the same conserved domain of IF proteins binds to the C-terminus of desmoplakin, which may help explain the previously reported broad binding IF-specificity to desmoplakin. Our data suggest that desmoplakin high-affinity binding to diverse IF proteins ensures robust linkages of IF cytoskeleton and desmosomes that maintain the structural integrity of cellular adhesion complexes. In summary, our results give new insights into the molecular basis of the IF-desmosome association.
Collapse
Affiliation(s)
- Bertrand Favre
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Nadja Begré
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Jamal-Eddine Bouameur
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Prakash Lingasamy
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Gloria M. Conover
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Lionel Fontao
- Department of Dermatology, Geneva University Hospitals, Geneva, Switzerland
| | - Luca Borradori
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
| |
Collapse
|
35
|
Aweida D, Rudesky I, Volodin A, Shimko E, Cohen S. GSK3-β promotes calpain-1-mediated desmin filament depolymerization and myofibril loss in atrophy. J Cell Biol 2018; 217:3698-3714. [PMID: 30061109 PMCID: PMC6168250 DOI: 10.1083/jcb.201802018] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 06/06/2018] [Accepted: 07/16/2018] [Indexed: 12/16/2022] Open
Abstract
Phosphorylation by protein kinase GSK3-β is essential for desmin filament depolymerization by calpain-1 and the resulting myofibril destruction in muscle atrophy. Myofibril breakdown is a fundamental cause of muscle wasting and inevitable sequel of aging and disease. We demonstrated that myofibril loss requires depolymerization of the desmin cytoskeleton, which is activated by phosphorylation. Here, we developed a mass spectrometry–based kinase-trap assay and identified glycogen synthase kinase 3-β (GSK3-β) as responsible for desmin phosphorylation. GSK3-β inhibition in mice prevented desmin phosphorylation and depolymerization and blocked atrophy upon fasting or denervation. Desmin was phosphorylated by GSK3-β 3 d after denervation, but depolymerized only 4 d later when cytosolic Ca2+ levels rose. Mass spectrometry analysis identified GSK3-β and the Ca2+-specific protease, calpain-1, bound to desmin and catalyzing its disassembly. Consistently, calpain-1 down-regulation prevented loss of phosphorylated desmin and blocked atrophy. Thus, phosphorylation of desmin filaments by GSK3-β is a key molecular event required for calpain-1–mediated depolymerization, and the subsequent myofibril destruction. Consequently, GSK3-β represents a novel drug target to prevent myofibril breakdown and atrophy.
Collapse
Affiliation(s)
- Dina Aweida
- Faculty of Biology, Technion Institute of Technology, Haifa, Israel
| | - Inga Rudesky
- Faculty of Biology, Technion Institute of Technology, Haifa, Israel
| | | | - Eitan Shimko
- Faculty of Biology, Technion Institute of Technology, Haifa, Israel
| | - Shenhav Cohen
- Faculty of Biology, Technion Institute of Technology, Haifa, Israel
| |
Collapse
|
36
|
Arginine methylation is required for canonical Wnt signaling and endolysosomal trafficking. Proc Natl Acad Sci U S A 2018; 115:E5317-E5325. [PMID: 29773710 PMCID: PMC6003351 DOI: 10.1073/pnas.1804091115] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Arginine methylation has emerged as a widespread and reversible protein modification with the potential to regulate a multitude of cellular processes, but its function is poorly understood. Endolysosomes play an important role in Wnt signaling, in which glycogen synthase kinase 3 (GSK3) becomes sequestered inside multivesicular bodies (MVBs) by the process known as microautophagy, causing the stabilization of many proteins. Up to 20% of cellular proteins contain three or more consecutive putative GSK3 sites, and of these 33% also contain methylarginine (meArg) modifications. Intriguingly, a cytoskeletal protein was previously known to have meArg modifications that enhanced subsequent phosphorylations by GSK3. Here, we report the unexpected finding that protein arginine methyltransferase 1 (PRMT1) is required for canonical Wnt signaling. Treatment of cultured cells for 5-30 min with Wnt3a induced a large increase in total endocytic vesicles which were also positive for asymmetric dimethylarginine modifications. Protease protection studies, both biochemical and in situ in cultured cells, showed that many meArg-modified cytosolic proteins became rapidly translocated into MVBs together with GSK3 and Lys48-polyubiquitinated proteins by ESCRT-driven microautophagy. In the case of the transcription factor Smad4, we showed that a unique arginine methylation site was required for GSK3 phosphorylation and Wnt regulation. The enzyme PRMT1 was found to be essential for Wnt-stimulated arginine methylation, GSK3 sequestration, and canonical Wnt signaling. The results reveal a cell biological role for PRMT1 arginine methylation at the crossroads of growth factor signaling, protein phosphorylation, membrane trafficking, cytosolic proteolysis, and Wnt-regulated microautophagy.
Collapse
|
37
|
Schlögl E, Radeva MY, Vielmuth F, Schinner C, Waschke J, Spindler V. Keratin Retraction and Desmoglein3 Internalization Independently Contribute to Autoantibody-Induced Cell Dissociation in Pemphigus Vulgaris. Front Immunol 2018; 9:858. [PMID: 29922278 PMCID: PMC5996934 DOI: 10.3389/fimmu.2018.00858] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/06/2018] [Indexed: 01/12/2023] Open
Abstract
Pemphigus vulgaris (PV) is a potentially lethal autoimmune disease characterized by blister formation of the skin and mucous membranes and is caused by autoantibodies against desmoglein (Dsg) 1 and Dsg3. Dsg1 and Dsg3 are linked to keratin filaments in desmosomes, adhering junctions abundant in tissues exposed to high levels of mechanical stress. The binding of the autoantibodies leads to internalization of Dsg3 and a collapse of the keratin cytoskeleton-yet, the relevance and interdependence of these changes for loss of cell-cell adhesion and blistering is poorly understood. In live-cell imaging studies, loss of the keratin network at the cell periphery was detectable starting after 60 min of incubation with immunoglobulin G fractions of PV patients (PV-IgG). These rapid changes correlated with loss of cell-cell adhesion detected by dispase-based dissociation assays and were followed by a condensation of keratin filaments into thick bundles after several hours. Dsg3 internalization started at 90 min of PV-IgG treatment, thus following the early keratin changes. By inhibiting casein kinase 1 (CK-1), we provoked keratin alterations resembling the effects of PV-IgG. Although CK-1-induced loss of peripheral keratin network correlated with loss of cell cohesion and Dsg3 clustering in the membrane, it was not sufficient to trigger the internalization of Dsg3. However, additional incubation with PV-IgG was effective to promote Dsg3 loss at the membrane, indicating that Dsg3 internalization is independent from keratin alterations. Vice versa, inhibiting Dsg3 internalization did not prevent PV-IgG-induced keratin retraction and only partially rescued cell cohesion. Together, keratin changes appear very early after autoantibody binding and temporally overlap with loss of cell cohesion. These early alterations appear to be distinct from Dsg3 internalization, suggesting a crucial role for initial loss of cell cohesion in PV.
Collapse
Affiliation(s)
- Elisabeth Schlögl
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Mariya Y Radeva
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Franziska Vielmuth
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Camilla Schinner
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Volker Spindler
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, Ludwig Maximilian University of Munich, Munich, Germany.,Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
38
|
Garcia MA, Nelson WJ, Chavez N. Cell-Cell Junctions Organize Structural and Signaling Networks. Cold Spring Harb Perspect Biol 2018; 10:a029181. [PMID: 28600395 PMCID: PMC5773398 DOI: 10.1101/cshperspect.a029181] [Citation(s) in RCA: 273] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell-cell junctions link cells to each other in tissues, and regulate tissue homeostasis in critical cell processes that include tissue barrier function, cell proliferation, and migration. Defects in cell-cell junctions give rise to a wide range of tissue abnormalities that disrupt homeostasis and are common in genetic abnormalities and cancers. Here, we discuss the organization and function of cell-cell junctions primarily involved in adhesion (tight junction, adherens junction, and desmosomes) in two different epithelial tissues: a simple epithelium (intestine) and a stratified epithelium (epidermis). Studies in these tissues reveal similarities and differences in the organization and functions of different cell-cell junctions that meet the requirements for the specialized functions of each tissue. We discuss cell-cell junction responses to genetic and environmental perturbations that provide further insights into their roles in maintaining tissue homeostasis.
Collapse
Affiliation(s)
- Miguel A Garcia
- Department of Biology, Stanford University, Stanford, California 94305
| | - W James Nelson
- Department of Biology, Stanford University, Stanford, California 94305
- Departments of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305
| | - Natalie Chavez
- Department of Biology, Stanford University, Stanford, California 94305
| |
Collapse
|
39
|
Hu L, Huang Z, Wu Z, Ali A, Qian A. Mammalian Plakins, Giant Cytolinkers: Versatile Biological Functions and Roles in Cancer. Int J Mol Sci 2018; 19:ijms19040974. [PMID: 29587367 PMCID: PMC5979291 DOI: 10.3390/ijms19040974] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/18/2018] [Accepted: 03/20/2018] [Indexed: 01/07/2023] Open
Abstract
Cancer is a highly lethal disease that is characterized by aberrant cell proliferation, migration, and adhesion, which are closely related to the dynamic changes of cytoskeletons and cytoskeletal-adhesion. These will further result in cell invasion and metastasis. Plakins are a family of giant cytolinkers that connect cytoskeletal elements with each other and to junctional complexes. With various isoforms composed of different domain structures, mammalian plakins are broadly expressed in numerous tissues. They play critical roles in many cellular processes, including cell proliferation, migration, adhesion, and signaling transduction. As these cellular processes are key steps in cancer development, mammalian plakins have in recent years attracted more and more attention for their potential roles in cancer. Current evidence shows the importance of mammalian plakins in various human cancers and demonstrates mammalian plakins as potential biomarkers for cancer. Here, we introduce the basic characteristics of mammalian plakins, review the recent advances in understanding their biological functions, and highlight their roles in human cancers, based on studies performed by us and others. This will provide researchers with a comprehensive understanding of mammalian plakins, new insights into the development of cancer, and novel targets for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Lifang Hu
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Zizhan Huang
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Zixiang Wu
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Arshad Ali
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Airong Qian
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| |
Collapse
|
40
|
Najor NA, Fitz GN, Koetsier JL, Godsel LM, Albrecht LV, Harmon R, Green KJ. Epidermal Growth Factor Receptor neddylation is regulated by a desmosomal-COP9 (Constitutive Photomorphogenesis 9) signalosome complex. eLife 2017; 6:22599. [PMID: 28891468 PMCID: PMC5663478 DOI: 10.7554/elife.22599] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 09/08/2017] [Indexed: 12/12/2022] Open
Abstract
Cell junctions are scaffolds that integrate mechanical and chemical signaling. We previously showed that a desmosomal cadherin promotes keratinocyte differentiation in an adhesion-independent manner by dampening Epidermal Growth Factor Receptor (EGFR) activity. Here we identify a potential mechanism by which desmosomes assist the de-neddylating COP9 signalosome (CSN) in attenuating EGFR through an association between the Cops3 subunit of the CSN and desmosomal components, Desmoglein1 (Dsg1) and Desmoplakin (Dp), to promote epidermal differentiation. Silencing CSN or desmosome components shifts the balance of EGFR modifications from ubiquitination to neddylation, inhibiting EGFR dynamics in response to an acute ligand stimulus. A reciprocal relationship between loss of Dsg1 and neddylated EGFR was observed in a carcinoma model, consistent with a role in sustaining EGFR activity during tumor progression. Identification of this previously unrecognized function of the CSN in regulating EGFR neddylation has broad-reaching implications for understanding how homeostasis is achieved in regenerating epithelia. The outer layer of skin – the epidermis – forms a critical barrier against a range of stresses from the environment. The epidermis itself consists of multiple layers of cells that are constantly being renewed. New cells are made in the deepest layer and move upwards until they eventually reach the skin’s surface. During this journey, the cells change the molecules they make in a process called epidermal differentiation. To maintain an effective barrier, the epidermis must balance the division of cells in the deepest layer with the differentiation of cells in the layers above. When activated, a protein called the Epidermal Growth Factor Receptor (or EGFR for short) encourages cells in the deepest layer to divide. However, it remains poorly understood how the balance between cells dividing and cells differentiating is achieved. The desmosome is a structure that can link together cells within the epidermis. Najor et al. now report a new interaction between the desmosome and a very large protein complex called the COP9- signalosome known to remove protein-based tags from other proteins. The experiments show that the COP9-signalosome results in the removal of these tags from EGFR. The status of the tags on EGFR regulates whether or not it is found at the cell surface. Najor et al. propose that that the desmosome acts as a scaffold and holds the COP9 signalosome close to EGFR. The enzyme in the COP9 signalosome then removes protein-based tags from EGFR, which triggers a series of events that remove EGFR from the cell surface. This dampens down the signals EGFR would normally send to make cells divide, and allows differentiation to proceed. The balance between cell division and differentiation is a fundamental process that is affected in many skin conditions, including psoriasis and atopic dermatitis. EGFR is also commonly overactive in cancers. As such, understanding how epidermal differentiation and cell division are controlled will shed light on a variety of disorders, allowing for the potential development of new treatments for these diseases.
Collapse
Affiliation(s)
- Nicole Ann Najor
- Department of Biology, College of Engineering and Science, University of Detroit Mercy, Detroit, United States.,Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Gillian Nicole Fitz
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Jennifer Leigh Koetsier
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Lisa Marie Godsel
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, United States.,Department of Dermatology Chicago, Feinberg School of Medicine, Northwestern University, Evanston, United States
| | - Lauren Veronica Albrecht
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Robert Harmon
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Kathleen Janee Green
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, United States.,Department of Dermatology Chicago, Feinberg School of Medicine, Northwestern University, Evanston, United States
| |
Collapse
|
41
|
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: 12.3] [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
|
42
|
Broussard JA, Yang R, Huang C, Nathamgari SSP, Beese AM, Godsel LM, Hegazy MH, Lee S, Zhou F, Sniadecki NJ, Green KJ, Espinosa HD. The desmoplakin-intermediate filament linkage regulates cell mechanics. Mol Biol Cell 2017; 28:3156-3164. [PMID: 28495795 PMCID: PMC5687018 DOI: 10.1091/mbc.e16-07-0520] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 03/16/2017] [Accepted: 05/02/2017] [Indexed: 02/06/2023] Open
Abstract
Desmoplakin connects desmosomal core components to intermediate filaments at sites of cell–cell adhesion. Modulating the strength of this linkage using desmoplakin mutants led to alterations in cell–substrate and cell–cell forces and cell stiffness as assessed by micropillar arrays and atomic force microscopy. Perturbation of the actin cytoskeleton leads to abrogation of these effects. The translation of mechanical forces into biochemical signals plays a central role in guiding normal physiological processes during tissue development and homeostasis. Interfering with this process contributes to cardiovascular disease, cancer progression, and inherited disorders. The actin-based cytoskeleton and its associated adherens junctions are well-established contributors to mechanosensing and transduction machinery; however, the role of the desmosome–intermediate filament (DSM–IF) network is poorly understood in this context. Because a force balance among different cytoskeletal systems is important to maintain normal tissue function, knowing the relative contributions of these structurally integrated systems to cell mechanics is critical. Here we modulated the interaction between DSMs and IFs using mutant forms of desmoplakin, the protein bridging these structures. Using micropillar arrays and atomic force microscopy, we demonstrate that strengthening the DSM–IF interaction increases cell–substrate and cell–cell forces and cell stiffness both in cell pairs and sheets of cells. In contrast, disrupting the interaction leads to a decrease in these forces. These alterations in cell mechanics are abrogated when the actin cytoskeleton is dismantled. These data suggest that the tissue-specific variability in DSM–IF network composition provides an opportunity to differentially regulate tissue mechanics by balancing and tuning forces among cytoskeletal systems.
Collapse
Affiliation(s)
- Joshua A Broussard
- Department of Pathology, Northwestern University, Chicago, IL 60611.,Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Ruiguo Yang
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208
| | - Changjin Huang
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208
| | - S Shiva P Nathamgari
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208
| | - Allison M Beese
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208
| | - Lisa M Godsel
- Department of Pathology, Northwestern University, Chicago, IL 60611.,Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Marihan H Hegazy
- Department of Pathology, Northwestern University, Chicago, IL 60611
| | - Sherry Lee
- Department of Pathology, Northwestern University, Chicago, IL 60611
| | - Fan Zhou
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208
| | - Nathan J Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195.,Department of Bioengineering, University of Washington, Seattle, WA 98195.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195
| | - Kathleen J Green
- Department of Pathology, Northwestern University, Chicago, IL 60611 .,Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Horacio D Espinosa
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208 .,Theoretical and Applied Mechanics Program, Northwestern University, Evanston, IL 60208
| |
Collapse
|
43
|
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: 28] [Impact Index Per Article: 4.0] [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
|
44
|
Onwuli DO, Rigau-Roca L, Cawthorne C, Beltran-Alvarez P. Mapping arginine methylation in the human body and cardiac disease. Proteomics Clin Appl 2016; 11. [PMID: 27600370 DOI: 10.1002/prca.201600106] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 08/20/2016] [Accepted: 09/02/2016] [Indexed: 01/11/2023]
Abstract
PURPOSE Arginine methylation (ArgMe) is one of the most ubiquitous PTMs, and hundreds of proteins undergo ArgMe in, for example, brain. However, the scope of ArgMe in many tissues, including the heart, is currently underexplored. Here, we aimed to (i) identify proteins undergoing ArgMe in human organs, and (ii) expose the relevance of ArgMe in cardiac disease. EXPERIMENTAL DESIGN The publicly available proteomic data is used to search for ArgMe in 13 human tissues. To induce H9c2 cardiac-like cell hypertrophy glucose is used. RESULTS The results show that ArgMe is mainly tissue-specific; nevertheless, the authors suggest an embryonic origin of core ArgMe events. In the heart, 103 mostly novel ArgMe sites in 58 nonhistone proteins are found. The authors provide compelling evidence that cardiac protein ArgMe is relevant to cardiomyocyte ontology, and important for proper cardiac function. This is highlighted by the fact that genetic mutations affecting methylated arginine positions are often associated with cardiac disease, including hypertrophic cardiomyopathy. The pilot experimental data suggesting significant changes in ArgMe profiles of H9c2 cells upon induction of cell hypertrophy using glucose is provided. CONCLUSIONS AND CLINICAL RELEVANCE The work calls for in-depth investigation of ArgMe in normal and diseased tissues using methods including clinical proteomics.
Collapse
|
45
|
Abstract
This review discusses the spectrin superfamily of proteins that function to connect cytoskeletal elements to each other, the cell membrane, and the nucleus. The signature domain is the spectrin repeat, a 106-122-amino-acid segment comprising three α-helices. α-actinin is considered to be the ancestral protein and functions to cross-link actin filaments. It then evolved to generate spectrin and dystrophin that function to link the actin cytoskeleton to the cell membrane, as well as the spectraplakins and plakins that link cytoskeletal elements to each other and to junctional complexes. A final class comprises the nesprins, which are able to bind to the nuclear membrane. This review discusses the domain organization of the various spectrin family members, their roles in protein-protein interactions, and their roles in disease, as determined from mutations, and it also describes the functional roles of the family members as determined from null phenotypes.
Collapse
Affiliation(s)
- Ronald K H Liem
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York 10032
| |
Collapse
|
46
|
McAnany CE, Mura C. Claws, Disorder, and Conformational Dynamics of the C-Terminal Region of Human Desmoplakin. J Phys Chem B 2016; 120:8654-67. [PMID: 27188911 DOI: 10.1021/acs.jpcb.6b03261] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Multicellular organisms consist of cells that interact via elaborate adhesion complexes. Desmosomes are membrane-associated adhesion complexes that mechanically tether the cytoskeletal intermediate filaments (IFs) between two adjacent cells, creating a network of tough connections in tissues such as skin and heart. Desmoplakin (DP) is the key desmosomal protein that binds IFs, and the DP·IF association poses a quandary: desmoplakin must stably and tightly bind IFs to maintain the structural integrity of the desmosome. Yet, newly synthesized DP must traffic along the cytoskeleton to the site of nascent desmosome assembly without "sticking" to the IF network, implying weak or transient DP···IF contacts. Recent work reveals that these contacts are modulated by post-translational modifications (PTMs) in DP's C-terminal tail (DPCTT). Using molecular dynamics simulations, we have elucidated the structural basis of these PTM-induced effects. Our simulations, nearing 2 μs in aggregate, indicate that phosphorylation of S2849 induces an "arginine claw" in desmoplakin's C-terminal tail. If a key arginine, R2834, is methylated, the DPCTT preferentially samples conformations that are geometrically well-suited as substrates for processive phosphorylation by the cognate kinase GSK3. We suggest that DPCTT is a molecular switch that modulates, via its conformational dynamics, DP's overall efficacy as a substrate for GSK3. Finally, we show that the fluctuating DPCTT can contact other parts of DP, suggesting a competitive binding mechanism for the modulation of DP···IF interactions.
Collapse
Affiliation(s)
- Charles E McAnany
- Department of Chemistry, University of Virginia , Charlottesville, Virginia 22904, United States
| | - Cameron Mura
- Department of Chemistry, University of Virginia , Charlottesville, Virginia 22904, United States
| |
Collapse
|
47
|
|
48
|
Loschke F, Homberg M, Magin TM. Keratin Isotypes Control Desmosome Stability and Dynamics through PKCα. J Invest Dermatol 2016; 136:202-13. [DOI: 10.1038/jid.2015.403] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 09/04/2015] [Accepted: 09/11/2015] [Indexed: 02/08/2023]
|
49
|
Martherus R, Jain R, Takagi K, Mendsaikhan U, Turdi S, Osinska H, James JF, Kramer K, Purevjav E, Towbin JA. Accelerated cardiac remodeling in desmoplakin transgenic mice in response to endurance exercise is associated with perturbed Wnt/β-catenin signaling. Am J Physiol Heart Circ Physiol 2015; 310:H174-87. [PMID: 26545710 DOI: 10.1152/ajpheart.00295.2015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 10/02/2015] [Indexed: 12/13/2022]
Abstract
Arrhythmogenic ventricular cardiomyopathy (AVC) is a frequent underlying cause for arrhythmias and sudden cardiac death especially during intense exercise. The mechanisms involved remain largely unknown. The purpose of this study was to investigate how chronic endurance exercise contributes to desmoplakin (DSP) mutation-induced AVC pathogenesis. Transgenic mice with overexpression of desmoplakin, wild-type (Tg-DSP(WT)), or the R2834H mutant (Tg-DSP(R2834H)) along with control nontransgenic (NTg) littermates were kept sedentary or exposed to a daily running regimen for 12 wk. Cardiac function and morphology were analyzed using echocardiography, electrocardiography, histology, immunohistochemistry, RNA, and protein analysis. At baseline, 4-wk-old mice from all groups displayed normal cardiac function. When subjected to exercise, all mice retained normal cardiac function and left ventricular morphology; however, Tg-DSP(R2834H) mutants displayed right ventricular (RV) dilation and wall thinning, unlike NTg and Tg-DSP(WT). The Tg-DSP(R2834H) hearts demonstrated focal fat infiltrations in RV and cytoplasmic aggregations consisting of desmoplakin, plakoglobin, and connexin 43. These aggregates coincided with disruption of the intercalated disks, intermediate filaments, and microtubules. Although Tg-DSP(R2834H) mice already displayed high levels of p-GSK3-β(Ser9) and p-AKT1(Ser473) under sedentary conditions, decrease of nuclear GSK3-β and AKT1 levels with reduced p-GSK3-β(Ser9), p-AKT1(Ser473), and p-AKT1(Ser308) and loss of nuclear junctional plakoglobin was apparent after exercise. In contrast, Tg-DSP(WT) showed upregulation of p-AKT1(Ser473), p-AKT1(Ser308), and p-GSK3-β(Ser9) in response to exercise. Our data suggest that endurance exercise accelerates AVC pathogenesis in Tg-DSP(R2834H) mice and this event is associated with perturbed AKT1 and GSK3-β signaling. Our study suggests a potential mechanism-based approach to exercise management in patients with AVC.
Collapse
Affiliation(s)
- Ruben Martherus
- Cardiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Rahul Jain
- Department of Cardiology, Indiana University, Indianapolis, Indiana; and
| | - Ken Takagi
- Cardiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Jikei University, Tokyo, Japan
| | - Uzmee Mendsaikhan
- Cardiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Subat Turdi
- Cardiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Hanna Osinska
- Cardiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jeanne F James
- Cardiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kristen Kramer
- Cardiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Enkhsaikhan Purevjav
- Cardiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jeffrey A Towbin
- Cardiology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| |
Collapse
|
50
|
Functional Analysis of Periplakin and Envoplakin, Cytoskeletal Linkers, and Cornified Envelope Precursor Proteins. Methods Enzymol 2015; 569:309-29. [PMID: 26778565 DOI: 10.1016/bs.mie.2015.06.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Envoplakin and periplakin are the two smallest plakin family cytoskeletal linker proteins that connect intermediate filaments to cellular junctions and other membrane locations. These two plakins have a structural role in the assembly of the cornified envelope (CE), the terminal stage of epidermal differentiation. Analysis of gene-targeted mice lacking both these plakins and the third initial CE scaffold protein, involucrin, demonstrate the importance of the structural integrity of CE for a proper epidermal barrier function. It has emerged that periplakin, which also has a wider tissue distribution than envoplakin, has additional, independent roles. Periplakin participates in the cytoskeletal organization also in other tissues and interacts with a wide range of membrane-associated proteins such as kazrin and butyrophilin BTN3A1. This review covers methods used to understand periplakin and envoplakin functions in cell culture models, including siRNA ablation of periplakin expression and the use of tagged protein domain constructs to study localization and interactions. In addition, assays that can be used to analyze CEs and epidermal barrier function in gene-targeted mice are described and discussed.
Collapse
|