1
|
Ruiz WG, Clayton DR, Parakala-Jain T, Dalghi MG, Franks J, Apodaca G. The rat bladder umbrella cell keratin network: Organization, dependence on the plectin cytolinker, and responses to bladder filling. Mol Biol Cell 2024; 35:ar139. [PMID: 39356795 PMCID: PMC11617100 DOI: 10.1091/mbc.e24-06-0262] [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: 06/17/2024] [Revised: 09/11/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024] Open
Abstract
The keratin cytoskeleton and associated desmosomes contribute to the mechanical stability of epithelial tissues, but their organization in native bladder umbrella cells and their responses to bladder filling are poorly understood. Using whole rat bladders in conjunction with confocal microscopy, super-resolution image processing, three-dimensional image reconstruction, and platinum replica electron microscopy, we identified a cortical cytoskeleton network in umbrella cells that was organized as a dense tile-like mesh comprised of tesserae bordered by cortical actin filaments, filled with keratin filaments, and cross-linked by plectin. Below these tesserae, keratin formed a subapical meshwork and at the cell periphery a band of keratin was linked via plectin to the junction-associated actin ring. Disruption of plectin led to focal keratin network dissolution, loss of the junction-associated keratin, and defects in cell-cell adhesion. During bladder filling, a junction-localized necklace of desmosomes expanded, and a subjacent girded layer formed linking the keratin network to desmosomes, including those at the umbrella cell-intermediate cell interface. Our studies reveal a novel tile- and mesh-like organization of the umbrella cell keratin network that is dependent on plectin, that reorganizes in response to bladder filling, and that likely serves to maintain umbrella cell continuity in the face of mechanical distension.
Collapse
Affiliation(s)
- Wily G. Ruiz
- Department of Medicine Renal-Electrolyte Division and George M. O'Brien Pittsburgh Center for Kidney Research, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Dennis R. Clayton
- Department of Medicine Renal-Electrolyte Division and George M. O'Brien Pittsburgh Center for Kidney Research, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Tanmay Parakala-Jain
- Department of Medicine Renal-Electrolyte Division and George M. O'Brien Pittsburgh Center for Kidney Research, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Marianela G. Dalghi
- Department of Medicine Renal-Electrolyte Division and George M. O'Brien Pittsburgh Center for Kidney Research, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jonathan Franks
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh 15213
| | - Gerard Apodaca
- Department of Medicine Renal-Electrolyte Division and George M. O'Brien Pittsburgh Center for Kidney Research, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh 15213
| |
Collapse
|
2
|
Ruiz WG, Clayton DR, Parakala-Jain T, Dalghi MG, Franks J, Apodaca G. The umbrella cell keratin network: organization as a tile-like mesh, formation of a girded layer in response to bladder filling, and dependence on the plectin cytolinker. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598498. [PMID: 38915686 PMCID: PMC11195278 DOI: 10.1101/2024.06.11.598498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
The keratin cytoskeleton and associated desmosomes contribute to the mechanical stability of epithelial tissues, but their organization in bladder umbrella cells and their responses to bladder filling are poorly understood. Using super-resolution confocal microscopy, along with 3D image reconstruction and platinum replica electron microscopy, we observed that the apical keratin network of umbrella cells was organized as a dense tile-like mesh comprised of tesserae bordered on their edges by cortical actin filaments, filled with woven keratin filaments, and crosslinked by plectin. A band of keratin was also observed at the cell periphery that was linked to the junction-associated actin ring by plectin. During bladder filling, the junction-localized desmosomal necklace expanded, and a subjacent girded layer was formed that linked the keratin network to desmosomes, including those at the umbrella cell-intermediate cell interface. Disruption of plectin led to focal keratin network dissolution, loss of the junction-associated band of keratin, perturbation of tight junction continuity, and loss of cell-cell cohesion. Our studies reveal a novel tile-like organization of the umbrella cell keratin cytoskeleton that is dependent on plectin, that reorganizes in response to bladder filling, and that likely serves to maintain umbrella cell continuity in the face of mechanical distension.
Collapse
Affiliation(s)
- Wily G. Ruiz
- Department of Medicine Renal-Electrolyte Division and George M. O’Brien Pittsburgh Center for Kidney Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dennis R. Clayton
- Department of Medicine Renal-Electrolyte Division and George M. O’Brien Pittsburgh Center for Kidney Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tanmay Parakala-Jain
- Department of Medicine Renal-Electrolyte Division and George M. O’Brien Pittsburgh Center for Kidney Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Marianela G. Dalghi
- Department of Medicine Renal-Electrolyte Division and George M. O’Brien Pittsburgh Center for Kidney Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jonathan Franks
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Gerard Apodaca
- Department of Medicine Renal-Electrolyte Division and George M. O’Brien Pittsburgh Center for Kidney Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Myo Min KK, Ffrench CB, McClure BJ, Ortiz M, Dorward EL, Samuel MS, Ebert LM, Mahoney MG, Bonder CS. Desmoglein-2 as a cancer modulator: friend or foe? Front Oncol 2023; 13:1327478. [PMID: 38188287 PMCID: PMC10766750 DOI: 10.3389/fonc.2023.1327478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Desmoglein-2 (DSG2) is a calcium-binding single pass transmembrane glycoprotein and a member of the large cadherin family. Until recently, DSG2 was thought to only function as a cell adhesion protein embedded within desmosome junctions designed to enable cells to better tolerate mechanical stress. However, additional roles for DSG2 outside of desmosomes are continuing to emerge, particularly in cancer. Herein, we review the current literature on DSG2 in cancer and detail its impact on biological functions such as cell adhesion, proliferation, migration, invasion, intracellular signaling, extracellular vesicle release and vasculogenic mimicry. An increased understanding of the diverse repertoire of the biological functions of DSG2 holds promise to exploit this cell surface protein as a potential prognostic biomarker and/or target for better patient outcomes. This review explores the canonical and non-canonical functions of DSG2, as well as the context-dependent impacts of DSG2 in the realm of cancer.
Collapse
Affiliation(s)
- Kay K. Myo Min
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Charlie B. Ffrench
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Barbara J. McClure
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Michael Ortiz
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Emma L. Dorward
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Michael S. Samuel
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Basil Hetzel Institute, Queen Elizabeth Hospital, SA, Adelaide, Australia
| | - Lisa M. Ebert
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
- Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Mỹ G. Mahoney
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Claudine S. Bonder
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
4
|
Zhang B, Wu Y, Yang X, Xiang Y, Yang B. Molecular insight into arrhythmogenic cardiomyopathy caused by DSG2 mutations. Biomed Pharmacother 2023; 167:115448. [PMID: 37696084 DOI: 10.1016/j.biopha.2023.115448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/13/2023] Open
Abstract
Mutant desmoglein 2 (DSG2) is the second most common pathogenic gene in arrhythmogenic cardiomyopathy (ACM), accounting for approximately 10% of ACM cases. In addition to common clinical and pathological features, ACM caused by mutant DSG2 has specific characteristics, manifesting as left ventricle involvement and a high risk of heart failure. Pathological studies have shown extensive cardiomyocyte necrosis, infiltration of immune cells, and fibrofatty replacement in both ventricles, as well as abnormal desmosome structures in the hearts of humans and mice with mutant DSG2-related ACM. Although desmosome dysfunction is a common pathway in the pathogenesis of mutant DSG2-related ACM, the mechanisms underlying this dysfunction vary among mutations. Desmosome dysfunction induces cardiomyocyte injury, plakoglobin dislocation, and gap junction dysfunction, all of which contribute to the initiation and progression of ACM. Additionally, dysregulated inflammation, overactivation of transforming growth factor-beta-1 signaling and endoplasmic reticulum stress, and cardiac metabolic dysfunction contribute to the pathogenesis of ACM caused by mutant DSG2. These features demonstrate that patients with mutant DSG2-related ACM should be managed individually and precisely based on the genotype and phenotype. Further studies are needed to investigate the underlying mechanisms and to identify novel therapies to reverse or attenuate the progression of ACM caused by mutant DSG2.
Collapse
Affiliation(s)
- Baowei Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai 200120, PR China
| | - Yizhang Wu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai 200120, PR China
| | - Xingbo Yang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road, Pudong, Shanghai 200120, PR China
| | - Yaozu Xiang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road, Pudong, Shanghai 200120, PR China.
| | - Bing Yang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai 200120, PR China.
| |
Collapse
|
5
|
Dean WF, Mattheyses AL. Defining domain-specific orientational order in the desmosomal cadherins. Biophys J 2022; 121:4325-4341. [PMID: 36225113 PMCID: PMC9703042 DOI: 10.1016/j.bpj.2022.10.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 09/12/2022] [Accepted: 10/06/2022] [Indexed: 01/25/2023] Open
Abstract
Desmosomes are large, macromolecular protein assemblies that mechanically couple the intermediate filament cytoskeleton to sites of cadherin-mediated cell adhesion, thereby providing structural integrity to tissues that routinely experience large forces. Proper desmosomal adhesion is necessary for the normal development and maintenance of vertebrate tissues, such as epithelia and cardiac muscle, while dysfunction can lead to severe disease of the heart and skin. Therefore, it is important to understand the relationship between desmosomal adhesion and the architecture of the molecules that form the adhesive interface, the desmosomal cadherins (DCs). However, desmosomes are embedded in two plasma membranes and are linked to the cytoskeletal networks of two cells, imposing extreme difficulty on traditional structural studies of DC architecture, which have yielded conflicting results. Consequently, the relationship between DC architecture and adhesive function remains unclear. To overcome these challenges, we utilized excitation-resolved fluorescence polarization microscopy to quantify the orientational order of the extracellular and intracellular domains of three DC isoforms: desmoglein 2, desmocollin 2, and desmoglein 3. We found that DC ectodomains were significantly more ordered than their cytoplasmic counterparts, indicating a drastic difference in DC architecture between opposing sides of the plasma membrane. This difference was conserved among all DCs tested, suggesting that it may be an important feature of desmosomal architecture. Moreover, our findings suggest that the organization of DC ectodomains is predominantly the result of extracellular adhesive interactions. We employed azimuthal orientation mapping to show that DC ectodomains are arranged with rotational symmetry about the membrane normal. Finally, we performed a series of mathematical simulations to test the feasibility of a recently proposed antiparallel arrangement of DC ectodomains, finding that it is supported by our experimental data. Importantly, the strategies employed here have the potential to elucidate molecular mechanisms for diseases that result from defective desmosome architecture.
Collapse
Affiliation(s)
- William F Dean
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Alexa L Mattheyses
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
6
|
Yang T, Jia L, Bian S, Chang X, Zhang Q, Tang Q, Zhu J, Yang Z, Feng Z. TROP2 Down-Regulated DSG2 to Promote Gastric Cancer Cell Invasion and Migration by EGFR/AKT and DSG2/PG/β-Catenin Pathways. Curr Cancer Drug Targets 2022; 22:691-702. [PMID: 35392784 DOI: 10.2174/1568009622666220407111013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/01/2021] [Accepted: 12/20/2021] [Indexed: 11/22/2022]
Abstract
AIMS Explore the specific mechanism of TROP2 in promoting cancer in gastric cancer, and provide a basis for the prevention and treatment of gastric cancer. Background Gastric cancer (GC) is the fourth most commonly found cancer and the second highest cause of cancer related death worldwide, TROP2 overexpression is closely related with many cancers including gastrointestinal tumors, DSG2 is an important protein in cell adhesion and its loss is related to cell migration. OBJECTIVE Explore the specific mechanism of TROP2 in promoting cancer in gastric cancer, and provide a basis for the prevention and treatment of gastric cancer. METHOD DSG2 was identified as an interacting protein of TROP2 in GC cells by co-immunoprecipitation and mass spectrometry. The regulated behavior of TROP2 on DSG2 expression was investigated with TROP2 over-expressure or knockdown. Cell-cell adhesion capacity medicated by DSG2 was evaluated by adhesion related assays. Electron microscope observation was utilized for accessing GC tumor desmosome assembly. Proteins in EGFR/AKT and DSG2/PG/β-catenin pathways were evaluated by western blotting. RESULT This study suggests that abundant expression of TROP2 in GC cells lessened DSG2 levels as well as desmosome adhesion, increased cell invasion, migration and promoted malignant progression through EGFR/AKT and DSG2/PG/β-catenin pathways. CONCLUSION TROP2 promotes gastric cancer cell invasion and migration by decreasing DSG2 expression through EGFR/AKT and DSG2/PG/β-catenin pathways.
Collapse
Affiliation(s)
- Tingting Yang
- Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China.,Department of Pathology, Nanjing Medical University, Nanjing 211166, China
| | - Lizhou Jia
- Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China
| | - Susu Bian
- Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China.,Department of Pathology, Nanjing Medical University, Nanjing 211166, China
| | - Xinxia Chang
- Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China
| | - Qian Zhang
- Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China
| | - Qi Tang
- Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China.,Department of Pathology, Nanjing Medical University, Nanjing 211166, China
| | - Jing Zhu
- Huadong Medical Institute of Biotechniques, Nanjing 210000, China
| | - Zhiping Yang
- Cancer Center, Bayannur Hospital, Bayannur, Inner Mongolia 015000, China
| | - Zhenqing Feng
- Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China.,Department of Pathology, Nanjing Medical University, Nanjing 211166, China.,Jiangsu Key Lab. of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
7
|
Hawthorne RN, Blazeski A, Lowenthal J, Kannan S, Teuben R, DiSilvestre D, Morrissette-McAlmon J, Saffitz JE, Boheler KR, James CA, Chelko SP, Tomaselli G, Tung L. Altered Electrical, Biomolecular, and Immunologic Phenotypes in a Novel Patient-Derived Stem Cell Model of Desmoglein-2 Mutant ARVC. J Clin Med 2021; 10:jcm10143061. [PMID: 34300226 PMCID: PMC8306340 DOI: 10.3390/jcm10143061] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 12/27/2022] Open
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a progressive heart condition which causes fibro-fatty myocardial scarring, ventricular arrhythmias, and sudden cardiac death. Most cases of ARVC can be linked to pathogenic mutations in the cardiac desmosome, but the pathophysiology is not well understood, particularly in early phases when arrhythmias can develop prior to structural changes. Here, we created a novel human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) model of ARVC from a patient with a c.2358delA variant in desmoglein-2 (DSG2). These DSG2-mutant (DSG2Mut) hiPSC-CMs were compared against two wildtype hiPSC-CM lines via immunostaining, RT-qPCR, Western blot, RNA-Seq, cytokine expression and optical mapping. Mutant cells expressed reduced DSG2 mRNA and had altered localization of desmoglein-2 protein alongside thinner, more disorganized myofibrils. No major changes in other desmosomal proteins were noted. There was increased pro-inflammatory cytokine expression that may be linked to canonical and non-canonical NFκB signaling. Action potentials in DSG2Mut CMs were shorter with increased upstroke heterogeneity, while time-to-peak calcium and calcium decay rate were reduced. These were accompanied by changes in ion channel and calcium handling gene expression. Lastly, suppressing DSG2 in control lines via siRNA allowed partial recapitulation of electrical anomalies noted in DSG2Mut cells. In conclusion, the aberrant cytoskeletal organization, cytokine expression, and electrophysiology found DSG2Mut hiPSC-CMs could underlie early mechanisms of disease manifestation in ARVC patients.
Collapse
Affiliation(s)
- Robert N. Hawthorne
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
- Medical Scientist Training Program, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Adriana Blazeski
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
| | - Justin Lowenthal
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
- Medical Scientist Training Program, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Suraj Kannan
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
- Medical Scientist Training Program, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Roald Teuben
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
| | - Deborah DiSilvestre
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (D.D.); (C.A.J.)
| | - Justin Morrissette-McAlmon
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
| | - Jeffrey E. Saffitz
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
| | - Kenneth R. Boheler
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (D.D.); (C.A.J.)
| | - Cynthia A. James
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (D.D.); (C.A.J.)
| | - Stephen P. Chelko
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (D.D.); (C.A.J.)
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
- Correspondence: (S.P.C.); (G.T.); (L.T.); Tel.: +1-850-644-2215 (S.P.C.); +1-718-430-2801 (G.T.); +1-410-955-9603 (L.T.)
| | - Gordon Tomaselli
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (D.D.); (C.A.J.)
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Correspondence: (S.P.C.); (G.T.); (L.T.); Tel.: +1-850-644-2215 (S.P.C.); +1-718-430-2801 (G.T.); +1-410-955-9603 (L.T.)
| | - Leslie Tung
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (R.N.H.); (A.B.); (J.L.); (S.K.); (R.T.); (J.M.-M.); (K.R.B.)
- Correspondence: (S.P.C.); (G.T.); (L.T.); Tel.: +1-850-644-2215 (S.P.C.); +1-718-430-2801 (G.T.); +1-410-955-9603 (L.T.)
| |
Collapse
|
8
|
Myasnikov R, Brodehl A, Meshkov A, Kulikova O, Kiseleva A, Pohl GM, Sotnikova E, Divashuk M, Klimushina M, Zharikova A, Pokrovskaya M, Koretskiy S, Kharlap M, Mershina E, Sinitsyn V, Basargina E, Gandaeva L, Barskiy V, Boytsov S, Milting H, Drapkina O. The Double Mutation DSG2-p.S363X and TBX20-p.D278X Is Associated with Left Ventricular Non-Compaction Cardiomyopathy: Case Report. Int J Mol Sci 2021; 22:ijms22136775. [PMID: 34202524 PMCID: PMC8268202 DOI: 10.3390/ijms22136775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/10/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
Left ventricular non-compaction cardiomyopathy (LVNC) is a rare heart disease, with or without left ventricular dysfunction, which is characterized by a two-layer structure of the myocardium and an increased number of trabeculae. The study of familial forms of LVNC is helpful for risk prediction and genetic counseling of relatives. Here, we present a family consisting of three members with LVNC. Using a next-generation sequencing approach a combination of two (likely) pathogenic nonsense mutations DSG2-p.S363X and TBX20-p.D278X was identified in all three patients. TBX20 encodes the cardiac T-box transcription factor 20. DSG2 encodes desmoglein–2, which is part of the cardiac desmosomes and belongs to the cadherin family. Since the identified nonsense variant (DSG2-p.S363X) is localized in the extracellular domain of DSG2, we performed in vitro cell transfection experiments. These experiments revealed the absence of truncated DSG2 at the plasma membrane, supporting the pathogenic relevance of DSG2-p.S363X. In conclusion, we suggest that in the future, these findings might be helpful for genetic screening and counseling of patients with LVNC.
Collapse
Affiliation(s)
- Roman Myasnikov
- National Research Center for Therapy and Preventive Medicine, Petroverigskiy Lane 10, 101990 Moscow, Russia; (R.M.); (A.M.); (O.K.); (E.S.); (M.D.); (M.K.); (A.Z.); (M.P.); (S.K.); (M.K.); (O.D.)
| | - Andreas Brodehl
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, 32545 Bad Oeynhausen, Germany; (G.M.P.); (H.M.)
- Correspondence: (A.B.); (A.K.)
| | - Alexey Meshkov
- National Research Center for Therapy and Preventive Medicine, Petroverigskiy Lane 10, 101990 Moscow, Russia; (R.M.); (A.M.); (O.K.); (E.S.); (M.D.); (M.K.); (A.Z.); (M.P.); (S.K.); (M.K.); (O.D.)
| | - Olga Kulikova
- National Research Center for Therapy and Preventive Medicine, Petroverigskiy Lane 10, 101990 Moscow, Russia; (R.M.); (A.M.); (O.K.); (E.S.); (M.D.); (M.K.); (A.Z.); (M.P.); (S.K.); (M.K.); (O.D.)
| | - Anna Kiseleva
- National Research Center for Therapy and Preventive Medicine, Petroverigskiy Lane 10, 101990 Moscow, Russia; (R.M.); (A.M.); (O.K.); (E.S.); (M.D.); (M.K.); (A.Z.); (M.P.); (S.K.); (M.K.); (O.D.)
- Correspondence: (A.B.); (A.K.)
| | - Greta Marie Pohl
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, 32545 Bad Oeynhausen, Germany; (G.M.P.); (H.M.)
| | - Evgeniia Sotnikova
- National Research Center for Therapy and Preventive Medicine, Petroverigskiy Lane 10, 101990 Moscow, Russia; (R.M.); (A.M.); (O.K.); (E.S.); (M.D.); (M.K.); (A.Z.); (M.P.); (S.K.); (M.K.); (O.D.)
| | - Mikhail Divashuk
- National Research Center for Therapy and Preventive Medicine, Petroverigskiy Lane 10, 101990 Moscow, Russia; (R.M.); (A.M.); (O.K.); (E.S.); (M.D.); (M.K.); (A.Z.); (M.P.); (S.K.); (M.K.); (O.D.)
- All-Russia Research Institute of Agricultural Biotechnology, Timiryazevskaya Street, 42, 127550 Moscow, Russia
| | - Marina Klimushina
- National Research Center for Therapy and Preventive Medicine, Petroverigskiy Lane 10, 101990 Moscow, Russia; (R.M.); (A.M.); (O.K.); (E.S.); (M.D.); (M.K.); (A.Z.); (M.P.); (S.K.); (M.K.); (O.D.)
| | - Anastasia Zharikova
- National Research Center for Therapy and Preventive Medicine, Petroverigskiy Lane 10, 101990 Moscow, Russia; (R.M.); (A.M.); (O.K.); (E.S.); (M.D.); (M.K.); (A.Z.); (M.P.); (S.K.); (M.K.); (O.D.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Lomonosovsky Prospect 27, Building 10, 119991 Moscow, Russia
| | - Maria Pokrovskaya
- National Research Center for Therapy and Preventive Medicine, Petroverigskiy Lane 10, 101990 Moscow, Russia; (R.M.); (A.M.); (O.K.); (E.S.); (M.D.); (M.K.); (A.Z.); (M.P.); (S.K.); (M.K.); (O.D.)
| | - Sergey Koretskiy
- National Research Center for Therapy and Preventive Medicine, Petroverigskiy Lane 10, 101990 Moscow, Russia; (R.M.); (A.M.); (O.K.); (E.S.); (M.D.); (M.K.); (A.Z.); (M.P.); (S.K.); (M.K.); (O.D.)
| | - Maria Kharlap
- National Research Center for Therapy and Preventive Medicine, Petroverigskiy Lane 10, 101990 Moscow, Russia; (R.M.); (A.M.); (O.K.); (E.S.); (M.D.); (M.K.); (A.Z.); (M.P.); (S.K.); (M.K.); (O.D.)
| | - Elena Mershina
- Medical Research and Educational Center, Lomonosov Moscow State University, Lomonosovsky Prospect 27, Building 10, 119991 Moscow, Russia; (E.M.); (V.S.)
| | - Valentin Sinitsyn
- Medical Research and Educational Center, Lomonosov Moscow State University, Lomonosovsky Prospect 27, Building 10, 119991 Moscow, Russia; (E.M.); (V.S.)
| | - Elena Basargina
- National Medical Research Center for Children’s Health, Lomonosovsky Prospect 2, Building 1, 119991 Moscow, Russia; (E.B.); (L.G.); (V.B.)
| | - Leila Gandaeva
- National Medical Research Center for Children’s Health, Lomonosovsky Prospect 2, Building 1, 119991 Moscow, Russia; (E.B.); (L.G.); (V.B.)
| | - Vladimir Barskiy
- National Medical Research Center for Children’s Health, Lomonosovsky Prospect 2, Building 1, 119991 Moscow, Russia; (E.B.); (L.G.); (V.B.)
| | - Sergey Boytsov
- National Medical Research Center for Cardiology, 3-ya Cherepkovskaya Street, 15A, 121552 Moscow, Russia;
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, 32545 Bad Oeynhausen, Germany; (G.M.P.); (H.M.)
| | - Oxana Drapkina
- National Research Center for Therapy and Preventive Medicine, Petroverigskiy Lane 10, 101990 Moscow, Russia; (R.M.); (A.M.); (O.K.); (E.S.); (M.D.); (M.K.); (A.Z.); (M.P.); (S.K.); (M.K.); (O.D.)
| |
Collapse
|
9
|
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: 2.5] [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
|
10
|
Stahley SN, Basta LP, Sharan R, Devenport D. Celsr1 adhesive interactions mediate the asymmetric organization of planar polarity complexes. eLife 2021; 10:e62097. [PMID: 33529151 PMCID: PMC7857726 DOI: 10.7554/elife.62097] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 01/23/2021] [Indexed: 12/31/2022] Open
Abstract
To orchestrate collective polarization across tissues, planar cell polarity (PCP) proteins localize asymmetrically to cell junctions, a conserved feature of PCP that requires the atypical cadherin Celsr1. We report that mouse Celsr1 engages in both trans- and cis-interactions, and organizes into dense and highly stable punctate assemblies. We provide evidence suggesting that PCP-mutant variant of Celsr1, Celsr1Crsh, selectively impairs lateral cis-interactions. Although Celsr1Crsh mediates cell adhesion in trans, it displays increased mobility, diminishes junctional enrichment, and fails to engage in homophilic adhesion with the wild-type protein, phenotypes that can be rescued by ectopic cis-dimerization. Using biochemical and super-resolution microscopy approaches, we show that although Celsr1Crsh physically interacts with PCP proteins Frizzled6 and Vangl2, it fails to organize these proteins into asymmetric junctional complexes. Our results suggest mammalian Celsr1 functions not only as a trans-adhesive homodimeric bridge, but also as an organizer of intercellular Frizzled6 and Vangl2 asymmetry through lateral, cis-interactions.
Collapse
Affiliation(s)
- Sara N Stahley
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | - Lena P Basta
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | - Rishabh Sharan
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | - Danelle Devenport
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| |
Collapse
|
11
|
Abstract
Intercalated discs (ICDs) are highly orchestrated structures that connect neighboring cardiomyocytes in the heart. Three major complexes are distinguished in ICD: desmosome, adherens junction (AJ), and gap junction (GJ). Desmosomes are major cell adhesion junctions that anchor cell membrane to the intermediate filament network; AJs connect the actin cytoskeleton of adjacent cells; and gap junctions metabolically and electrically connect the cytoplasm of adjacent cardiomyocytes. All these complexes work as a single unit, the so-called area composita, interdependently rather than individually. Mutation or altered expression of ICD proteins results in various cardiac diseases, such as ARVC (arrhythmogenic right ventricular cardiomyopathy), dilated cardiomyopathy, and hypotrophy cardiomyopathy, eventually leading to heart failure. In this article, we first review the recent findings on the structural organization of ICD and their functions and then focus on the recent advances in molecular pathogenesis of the ICD-related heart diseases, which include two major areas: i) the ICD gene mutations in cardiac diseases, and ii) the involvement of ICD proteins in signal transduction pathways leading to myocardium remodeling and eventual heart failure. These major ICD-related signaling pathways include Wnt/β-catenin pathway, p38 MAPK cascade, Rho-dependent serum response factor (SRF) signaling, calcineurin/NFAT signaling, Hippo kinase cascade, etc., which are differentially regulated in pathological conditions.
Collapse
|
12
|
Zimmer SE, Kowalczyk AP. The desmosome as a model for lipid raft driven membrane domain organization. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183329. [PMID: 32376221 DOI: 10.1016/j.bbamem.2020.183329] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 01/14/2023]
Abstract
Desmosomes are cadherin-based adhesion structures that mechanically couple the intermediate filament cytoskeleton of adjacent cells to confer mechanical stress resistance to tissues. We have recently described desmosomes as mesoscale lipid raft membrane domains that depend on raft dynamics for assembly, function, and disassembly. Lipid raft microdomains are regions of the plasma membrane enriched in sphingolipids and cholesterol. These domains participate in membrane domain heterogeneity, signaling and membrane trafficking. Cellular structures known to be dependent on raft dynamics include the post-synaptic density in neurons, the immunological synapse, and intercellular junctions, including desmosomes. In this review, we discuss the current state of the desmosome field and put forward new hypotheses for the role of lipid rafts in desmosome adhesion, signaling and epidermal homeostasis. Furthermore, we propose that differential lipid raft affinity of intercellular junction proteins is a central driving force in the organization of the epithelial apical junctional complex.
Collapse
Affiliation(s)
- Stephanie E Zimmer
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University, Atlanta, GA 30322, United States of America; Department of Cell Biology, Emory University, Atlanta, GA 30322, United States of America
| | - Andrew P Kowalczyk
- Department of Cell Biology, Emory University, Atlanta, GA 30322, United States of America; Department of Dermatology, Emory University, Atlanta, GA 30322, United States of America.
| |
Collapse
|
13
|
Distal myopathy induced arrhythmogenic right ventricular cardiomyopathy in a pedigree carrying novel DSG2 null variant. Int J Cardiol 2020; 298:25-31. [DOI: 10.1016/j.ijcard.2019.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/14/2019] [Accepted: 10/02/2019] [Indexed: 01/15/2023]
|
14
|
A founder homozygous DSG2 variant in East Asia results in ARVC with full penetrance and heart failure phenotype. Int J Cardiol 2018; 274:263-270. [PMID: 30454721 DOI: 10.1016/j.ijcard.2018.06.105] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/17/2018] [Accepted: 06/27/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Variants in the desmoglein-2 (DSG2) gene account for a significant proportion of patients with Arrhythmogenic Right Ventricular Cardiomyopathy (ARVC). The aim of this study was to evaluate the genetic epidemiology of DSG2 and the impact of a frequent homozygous DSG2 variant in East Asia. METHODS Genetic screening of 14 ARVC related genes was performed in 118 unrelated index patients using next-generation sequencing. Following that, family screening, clinical evaluation and haplotype analysis were performed among eight probands who carry the same homozygous DSG2 variant. We also examined the histopathology and protein expression using immunofluorescence staining on the myocardial tissue of two probands undergoing heart transplant. RESULTS Eighteen (15.2%) patients bear rare putatively deleterious variants in DSG2, among which 8 patients shared the homozygous DSG2 p.Phe531Cys variant. Family screening demonstrated that only homozygous variant carriers exhibited definite ARVC phenotype with 100% penetrance, while heterozygous variant carriers were either unaffected or only presented mild ARVC related symptoms in 25% relatives. Left ventricular involvement and bi-ventricular failure were common among homozygous p. Phe531Cys variant patients even at early age. Haplotype analysis demonstrated p. Phe531Cys was a founder variant in East Asia population with an allele frequency of 0.12%. CONCLUSIONS We identified, for the first time, a homozygous founder variant of DSG2 in East Asia, which was at surprisingly high frequency of 8.47% among Chinese ARVC patients with a full penetrance. This result suggested an urgent demand of genetic counseling for the probands and their relatives with heterozygous variant.
Collapse
|
15
|
Ventrella R, Kaplan N, Hoover P, Perez White BE, Lavker RM, Getsios S. EphA2 Transmembrane Domain Is Uniquely Required for Keratinocyte Migration by Regulating Ephrin-A1 Levels. J Invest Dermatol 2018; 138:2133-2143. [PMID: 29705292 DOI: 10.1016/j.jid.2018.04.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 12/19/2022]
Abstract
EphA2 receptor tyrosine kinase is activated by ephrin-A1 ligand, which harbors a glycosylphosphatidylinositol anchor that enhances lipid raft localization. Although EphA2 and ephrin-A1 modulate keratinocyte migration and differentiation, the ability of this cell-cell communication complex to localize to different membrane regions in keratinocytes remains unknown. Using a combination of biochemical and imaging approaches, we provide evidence that ephrin-A1 and a ligand-activated form of EphA2 partition outside of lipid raft domains in response to calcium-mediated cell-cell contact stabilization in normal human epidermal keratinocytes. EphA2 transmembrane domain swapping with a shorter and molecularly distinct transmembrane domain of EphA1 resulted in decreased localization of this receptor tyrosine kinase at cell-cell junctions and increased expression of ephrin-A1, which is a negative regulator of keratinocyte migration. Accordingly, altered EphA2 membrane distribution at cell-cell contacts limited the ability of keratinocytes to seal linear scratch wounds in vitro in an ephrin-A1-dependent manner. Collectively, these studies highlight a key role for the EphA2 transmembrane domain in receptor-ligand membrane distribution at cell-cell contacts that modulates ephrin-A1 levels to allow for efficient keratinocyte migration with relevance for cutaneous wound healing.
Collapse
Affiliation(s)
- Rosa Ventrella
- Department of Dermatology, 303 East Chicago Avenue, Ward 9, Northwestern University, Chicago, Illinois, USA
| | - Nihal Kaplan
- Department of Dermatology, 303 East Chicago Avenue, Ward 9, Northwestern University, Chicago, Illinois, USA
| | - Paul Hoover
- Department of Dermatology, 303 East Chicago Avenue, Ward 9, Northwestern University, Chicago, Illinois, USA
| | - Bethany E Perez White
- Department of Dermatology, 303 East Chicago Avenue, Ward 9, Northwestern University, Chicago, Illinois, USA
| | - Robert M Lavker
- Department of Dermatology, 303 East Chicago Avenue, Ward 9, Northwestern University, Chicago, Illinois, USA
| | - Spiro Getsios
- Department of Dermatology, 303 East Chicago Avenue, Ward 9, Northwestern University, Chicago, Illinois, USA.
| |
Collapse
|
16
|
Nekrasova O, Harmon RM, Broussard JA, Koetsier JL, Godsel LM, Fitz GN, Gardel ML, Green KJ. Desmosomal cadherin association with Tctex-1 and cortactin-Arp2/3 drives perijunctional actin polymerization to promote keratinocyte delamination. Nat Commun 2018; 9:1053. [PMID: 29535305 PMCID: PMC5849617 DOI: 10.1038/s41467-018-03414-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/09/2018] [Indexed: 12/22/2022] Open
Abstract
The epidermis is a multi-layered epithelium that serves as a barrier against water loss and environmental insults. Its morphogenesis occurs through a tightly regulated program of biochemical and architectural changes during which basal cells commit to differentiate and move towards the skin's surface. Here, we reveal an unexpected role for the vertebrate cadherin desmoglein 1 (Dsg1) in remodeling the actin cytoskeleton to promote the transit of basal cells into the suprabasal layer through a process of delamination, one mechanism of epidermal stratification. Actin remodeling requires the interaction of Dsg1 with the dynein light chain, Tctex-1 and the actin scaffolding protein, cortactin. We demonstrate that Tctex-1 ensures the correct membrane compartmentalization of Dsg1-containing desmosomes, allowing cortactin/Arp2/3-dependent perijunctional actin polymerization and decreasing tension at E-cadherin junctions to promote keratinocyte delamination. Moreover, Dsg1 is sufficient to enable simple epithelial cells to exit a monolayer to form a second layer, highlighting its morphogenetic potential.
Collapse
Affiliation(s)
- Oxana Nekrasova
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, 60611, IL, USA
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, 60611, IL, USA
| | - Robert M Harmon
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, 60611, IL, USA
- Institute for Biophysical Dynamics, University of Chicago, Chicago, 60637, IL, USA
| | - Joshua A Broussard
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, 60611, IL, USA
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, 60611, IL, USA
| | - Jennifer L Koetsier
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, 60611, IL, USA
| | - Lisa M Godsel
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, 60611, IL, USA
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, 60611, IL, USA
| | - Gillian N Fitz
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, 60611, IL, USA
| | - Margaret L Gardel
- Institute for Biophysical Dynamics, University of Chicago, Chicago, 60637, IL, USA
| | - Kathleen J Green
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, 60611, IL, USA.
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, 60611, IL, USA.
| |
Collapse
|
17
|
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: 1.4] [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
|
18
|
Vielmuth F, Wanuske MT, Radeva MY, Hiermaier M, Kugelmann D, Walter E, Buechau F, Magin TM, Waschke J, Spindler V. Keratins Regulate the Adhesive Properties of Desmosomal Cadherins through Signaling. J Invest Dermatol 2017; 138:121-131. [PMID: 28899688 DOI: 10.1016/j.jid.2017.08.033] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 07/10/2017] [Accepted: 08/06/2017] [Indexed: 11/17/2022]
Abstract
Tightly controlled intercellular adhesion is crucial for the integrity and function of the epidermis. The keratin filament cytoskeleton anchors desmosomes, supramolecular complexes required for strong intercellular adhesion. We tested whether keratin filaments control cell adhesion by regulating the adhesive properties of desmosomal cadherins such as desmoglein (Dsg) 3. Atomic force microscopy and fluorescence recovery after photobleaching experiments showed reduced Dsg3 adhesive forces and membrane stability in murine keratinocytes lacking all keratin filaments. Impairment of the actin cytoskeleton also resulted in decreased Dsg3 immobilization but did not affect Dsg3 binding properties, indicating that the latter are exclusively controlled by keratins. Reduced binding forces were dependent on p38 mitogen-activated protein kinase activity, which was deregulated in keratin-deficient cells. In contrast, inhibition of protein kinase C signaling, which is known to be controlled by keratins, promoted and spatially stabilized Dsg3-mediated interactions in the membrane. These results show a previously unreported mechanism for how keratins stabilize intercellular adhesion on the level of single desmosomal adhesion molecules.
Collapse
Affiliation(s)
- Franziska Vielmuth
- Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Marie-Therès Wanuske
- Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Mariya Y Radeva
- Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Matthias Hiermaier
- Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Daniela Kugelmann
- Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Elias Walter
- Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Fanny Buechau
- Institute of Biology and Translational Center for Regenerative Medicine, Sächsischer Inkubator für klinische Translation, University of Leipzig, Leipzig, Germany
| | - Thomas M Magin
- Institute of Biology and Translational Center for Regenerative Medicine, Sächsischer Inkubator für klinische Translation, University of Leipzig, Leipzig, Germany
| | - Jens Waschke
- Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany.
| | - Volker Spindler
- Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Munich, Germany.
| |
Collapse
|
19
|
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: 6.8] [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
|
20
|
Roberts BJ, Svoboda RA, Overmiller AM, Lewis JD, Kowalczyk AP, Mahoney MG, Johnson KR, Wahl JK. Palmitoylation of Desmoglein 2 Is a Regulator of Assembly Dynamics and Protein Turnover. J Biol Chem 2016; 291:24857-24865. [PMID: 27703000 DOI: 10.1074/jbc.m116.739458] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/08/2016] [Indexed: 01/09/2023] Open
Abstract
Desmosomes are prominent adhesive junctions present between many epithelial cells as well as cardiomyocytes. The mechanisms controlling desmosome assembly and remodeling in epithelial and cardiac tissue are poorly understood. We recently identified protein palmitoylation as a mechanism regulating desmosome dynamics. In this study, we have focused on the palmitoylation of the desmosomal cadherin desmoglein-2 (Dsg2) and characterized the role that palmitoylation of Dsg2 plays in its localization and stability in cultured cells. We identified two cysteine residues in the juxtamembrane (intracellular anchor) domain of Dsg2 that, when mutated, eliminate its palmitoylation. These cysteine residues are conserved in all four desmoglein family members. Although mutant Dsg2 localizes to endogenous desmosomes, there is a significant delay in its incorporation into junctions, and the mutant is also present in a cytoplasmic pool. Triton X-100 solubility assays demonstrate that mutant Dsg2 is more soluble than wild-type protein. Interestingly, trafficking of the mutant Dsg2 to the cell surface was delayed, and a pool of the non-palmitoylated Dsg2 co-localized with lysosomal markers. Taken together, these data suggest that palmitoylation of Dsg2 regulates protein transport to the plasma membrane. Modulation of the palmitoylation status of desmosomal cadherins can affect desmosome dynamics.
Collapse
Affiliation(s)
- Brett J Roberts
- From the Department of Oral Biology, College of Dentistry, University of Nebraska Medical Center, Lincoln, Nebraska 68583
| | - Robert A Svoboda
- From the Department of Oral Biology, College of Dentistry, University of Nebraska Medical Center, Lincoln, Nebraska 68583
| | - Andrew M Overmiller
- the Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, and
| | - Joshua D Lewis
- the Departments of Cell Biology and Dermatology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Andrew P Kowalczyk
- the Departments of Cell Biology and Dermatology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - My G Mahoney
- the Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, and
| | - Keith R Johnson
- From the Department of Oral Biology, College of Dentistry, University of Nebraska Medical Center, Lincoln, Nebraska 68583.,the Eppley Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska 68198
| | - James K Wahl
- From the Department of Oral Biology, College of Dentistry, University of Nebraska Medical Center, Lincoln, Nebraska 68583,
| |
Collapse
|
21
|
Völlner F, Ali J, Kurrle N, Exner Y, Eming R, Hertl M, Banning A, Tikkanen R. Loss of flotillin expression results in weakened desmosomal adhesion and Pemphigus vulgaris-like localisation of desmoglein-3 in human keratinocytes. Sci Rep 2016; 6:28820. [PMID: 27346727 PMCID: PMC4922016 DOI: 10.1038/srep28820] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 06/09/2016] [Indexed: 01/01/2023] Open
Abstract
Desmosomes are adhesion plaques that mediate cell-cell adhesion in many tissues, including the epidermis, and generate mechanical resistance to tissues. The extracellular domains of desmosomal cadherin proteins, desmogleins and desmocollins, are required for the interaction with cadherins of the neighbouring cells, whereas their cytoplasmic tails associate with cytoplasmic proteins which mediate connection to intermediate filaments. Disruption of desmosomal adhesion by mutations, autoantibodies or bacterial toxins results in severe human disorders of e.g. the skin and the heart. Despite the vital role of desmosomes in various tissues, the details of their molecular assembly are not clear. We here show that the two members of the flotillin protein family directly interact with the cytoplasmic tails of desmogleins. Depletion of flotillins in human keratinocytes results in weakened desmosomal adhesion and reduced expression of desmoglein-3, most likely due to a reduction in the desmosomal pool due to increased turnover. In the absence of flotillins, desmoglein-3 shows an altered localisation pattern in the cell-cell junctions of keratinocytes, which is highly similar to the localisation observed upon treatment with pemphigus vulgaris autoantibodies. Thus, our data show that flotillins, which have previously been connected to the classical cadherins, are also of importance for the desmosomal cell adhesion.
Collapse
Affiliation(s)
- Frauke Völlner
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Jawahir Ali
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Nina Kurrle
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Yvonne Exner
- Department of Dermatology and Allergology, Philipps University of Marburg, Baldingerstrasse, 35043 Marburg, Germany
| | - Rüdiger Eming
- Department of Dermatology and Allergology, Philipps University of Marburg, Baldingerstrasse, 35043 Marburg, Germany
| | - Michael Hertl
- Department of Dermatology and Allergology, Philipps University of Marburg, Baldingerstrasse, 35043 Marburg, Germany
| | - Antje Banning
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Ritva Tikkanen
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| |
Collapse
|
22
|
Cadwell CM, Jenkins PM, Bennett V, Kowalczyk AP. Ankyrin-G Inhibits Endocytosis of Cadherin Dimers. J Biol Chem 2015; 291:691-704. [PMID: 26574545 DOI: 10.1074/jbc.m115.648386] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Indexed: 01/26/2023] Open
Abstract
Dynamic regulation of endothelial cell adhesion is central to vascular development and maintenance. Furthermore, altered endothelial adhesion is implicated in numerous diseases. Therefore, normal vascular patterning and maintenance require tight regulation of endothelial cell adhesion dynamics. However, the mechanisms that control junctional plasticity are not fully understood. Vascular endothelial cadherin (VE-cadherin) is an adhesive protein found in adherens junctions of endothelial cells. VE-cadherin mediates adhesion through trans interactions formed by its extracellular domain. Trans binding is followed by cis interactions that laterally cluster the cadherin in junctions. VE-cadherin is linked to the actin cytoskeleton through cytoplasmic interactions with β- and α-catenin, which serve to increase adhesive strength. Furthermore, p120-catenin binds to the cytoplasmic tail of cadherin and stabilizes it at the plasma membrane. Here we report that induced cis dimerization of VE-cadherin inhibits endocytosis independent of both p120 binding and trans interactions. However, we find that ankyrin-G, a protein that links membrane proteins to the spectrin-actin cytoskeleton, associates with VE-cadherin and inhibits its endocytosis. Ankyrin-G inhibits VE-cadherin endocytosis independent of p120 binding. We propose a model in which ankyrin-G associates with and inhibits the endocytosis of VE-cadherin cis dimers. Our findings support a novel mechanism for regulation of VE-cadherin endocytosis through ankyrin association with cadherin engaged in lateral interactions.
Collapse
Affiliation(s)
- Chantel M Cadwell
- From the Biochemistry, Cell, and Developmental Biology Graduate Program
| | - Paul M Jenkins
- the Howard Hughes Medical Institute, Department of Biochemistry, Cell Biology, and Neurobiology, Duke University Medical Center, Durham, North Carolina 27710
| | - Vann Bennett
- the Howard Hughes Medical Institute, Department of Biochemistry, Cell Biology, and Neurobiology, Duke University Medical Center, Durham, North Carolina 27710
| | - Andrew P Kowalczyk
- Department of Cell Biology, Department of Dermatology, and Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322 and
| |
Collapse
|
23
|
Abstract
Desmosomes represent adhesive, spot-like intercellular junctions that in association with intermediate filaments mechanically link neighboring cells and stabilize tissue architecture. In addition to this structural function, desmosomes also act as signaling platforms involved in the regulation of cell proliferation, differentiation, migration, morphogenesis, and apoptosis. Thus, deregulation of desmosomal proteins has to be considered to contribute to tumorigenesis. Proteolytic fragmentation and downregulation of desmosomal cadherins and plaque proteins by transcriptional or epigenetic mechanisms were observed in different cancer entities suggesting a tumor-suppressive role. However, discrepant data in the literature indicate that context-dependent differences based on alternative intracellular, signal transduction lead to altered outcome. Here, modulation of Wnt/β-catenin signaling by plakoglobin or desmoplakin and of epidermal growth factor receptor signaling appears to be of special relevance. This review summarizes current evidence on how desmosomal proteins participate in carcinogenesis, and depicts the molecular mechanisms involved.
Collapse
Affiliation(s)
- Otmar Huber
- a Institute of Biochemistry II, Jena University Hospital, Friedrich-Schiller-University Jena , Nonnenplan 2-4, 07743 Jena , Germany.,b Center for Sepsis Control and Care, Jena University Hospital , Erlanger Allee 101, 07747 Jena , Germany
| | - Iver Petersen
- c Institute of Pathology, Jena University Hospital, Friedrich-Schiller-University Jena , Ziegelmühlenweg 1, 07743 Jena , Germany
| |
Collapse
|
24
|
Fujiwara M, Nagatomo A, Tsuda M, Obata S, Sakuma T, Yamamoto T, Suzuki ST. Desmocollin-2 alone forms functional desmosomal plaques, with the plaque formation requiring the juxtamembrane region and plakophilins. J Biochem 2015; 158:339-53. [DOI: 10.1093/jb/mvv048] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 04/09/2015] [Indexed: 01/04/2023] Open
|
25
|
Broussard JA, Getsios S, Green KJ. Desmosome regulation and signaling in disease. Cell Tissue Res 2015; 360:501-12. [PMID: 25693896 DOI: 10.1007/s00441-015-2136-5] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 01/21/2015] [Indexed: 01/10/2023]
Abstract
Desmosomes are cell-cell adhesive organelles with a well-known role in forming strong intercellular adhesion during embryogenesis and in adult tissues subject to mechanical stress, such as the heart and skin. More recently, desmosome components have also emerged as cell signaling regulators. Loss of expression or interference with the function of desmosome molecules results in diseases of the heart and skin and contributes to cancer progression. However, the underlying molecular mechanisms that result in inherited and acquired disorders remain poorly understood. To address this question, researchers are directing their studies towards determining the functions that occur inside and outside of the junctions and the extent to which functions are adhesion-dependent or independent. This review focuses on recent discoveries that provide insights into the role of desmosomes and desmosome components in cell signaling and disease; wherever possible, we address molecular functions within and outside of the adhesive structure.
Collapse
Affiliation(s)
- Joshua A Broussard
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | | | | |
Collapse
|
26
|
Vielmuth F, Hartlieb E, Kugelmann D, Waschke J, Spindler V. Atomic force microscopy identifies regions of distinct desmoglein 3 adhesive properties on living keratinocytes. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 11:511-20. [PMID: 25510735 DOI: 10.1016/j.nano.2014.10.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/30/2014] [Accepted: 10/19/2014] [Indexed: 11/30/2022]
Abstract
Desmosomes provide strong cell-cell adhesion which is crucial for the integrity of tissues such as the epidermis. However, nothing is known about the distribution and binding properties of desmosomal adhesion molecules on keratinocytes. Here we used atomic force microscopy (AFM) to simultaneously visualize the topography of living human keratinocytes and the distribution and binding properties of the desmosomal adhesion molecule desmoglein 3 (Dsg3). Using recombinant Dsg3 as sensor, binding events were detectable diffusely and in clusters on the cell surface and at areas of cell-cell contact. This was blocked by removing Ca(2+) and by addition of Dsg3-specific antibodies indicating homophilic Dsg3 binding. Binding forces of Dsg3 molecules were lower on the cell surface compared to areas of cell-cell contact. Our data for the first time directly demonstrate the occurrence of Dsg3 molecules outside of desmosomes and show that Dsg3 adhesive properties differ depending on their localization. From the clinical editor: Using atomic force microscopy in the study of keratinocytes, this study directly demonstrates the occurrence of desmoglein 3 molecules outside of desmosomes and reveales that the adhesive properties of these molecules do differ depending on their localization.
Collapse
Affiliation(s)
- Franziska Vielmuth
- Institute of Anatomy and Cell Biology, Department I, Ludwig-Maximilians-Universität Munich, Munich, Germany.
| | - Eva Hartlieb
- Institute of Anatomy and Cell Biology, Department I, Ludwig-Maximilians-Universität Munich, Munich, Germany.
| | - Daniela Kugelmann
- Institute of Anatomy and Cell Biology, Department I, Ludwig-Maximilians-Universität Munich, Munich, Germany.
| | - Jens Waschke
- Institute of Anatomy and Cell Biology, Department I, Ludwig-Maximilians-Universität Munich, Munich, Germany.
| | - Volker Spindler
- Institute of Anatomy and Cell Biology, Department I, Ludwig-Maximilians-Universität Munich, Munich, Germany.
| |
Collapse
|
27
|
Patel DM, Green KJ. Desmosomes in the Heart: A Review of Clinical and Mechanistic Analyses. ACTA ACUST UNITED AC 2014; 21:109-28. [DOI: 10.3109/15419061.2014.906533] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
28
|
Jiang K, Rankin CR, Nava P, Sumagin R, Kamekura R, Stowell SR, Feng M, Parkos CA, Nusrat A. Galectin-3 regulates desmoglein-2 and intestinal epithelial intercellular adhesion. J Biol Chem 2014; 289:10510-10517. [PMID: 24567334 DOI: 10.1074/jbc.m113.538538] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The desmosomal cadherins, desmogleins, and desmocollins mediate strong intercellular adhesion. Human intestinal epithelial cells express the desmoglein-2 isoform. A proteomic screen for Dsg2-associated proteins in intestinal epithelial cells identified a lectin referred to as galectin-3 (Gal3). Gal3 bound to N-linked β-galactosides in Dsg2 extracellular domain and co-sedimented with caveolin-1 in lipid rafts. Down-regulation of Gal3 protein or incubation with lactose, a galactose-containing disaccharide that competitively inhibits galectin binding to Dsg2, decreased intercellular adhesion in intestinal epithelial cells. In the absence of functional Gal3, Dsg2 protein was internalized from the plasma membrane and degraded in the proteasome. These results report a novel role of Gal3 in stabilizing a desmosomal cadherin and intercellular adhesion in intestinal epithelial cells.
Collapse
Affiliation(s)
- Kun Jiang
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322
| | - Carl R Rankin
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322
| | - Porfirio Nava
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322; Department of Physiology, Biophysics and Neuroscience, CINVESTAV IPN., Av. IPN 2508, Col. San Pedro Zacatenco, 07360 Ciudad de México, Distrito Federal, México
| | - Ronen Sumagin
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322
| | - Ryuta Kamekura
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322
| | - Sean R Stowell
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322
| | - Mingli Feng
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322
| | - Charles A Parkos
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322
| | - Asma Nusrat
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology and Mucosal Inflammation Research Unit, Emory University, Atlanta, Georgia 30322.
| |
Collapse
|
29
|
Waschke J, Spindler V. Desmosomes and Extradesmosomal Adhesive Signaling Contacts in Pemphigus. Med Res Rev 2014; 34:1127-45. [DOI: 10.1002/med.21310] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Jens Waschke
- Institute of Anatomy and Cell Biology, Department I; Ludwig-Maximilians-Universität (LMU) Munich; Pettenkoferstrasse 11 D-80336 Munich Germany
| | - Volker Spindler
- Institute of Anatomy and Cell Biology, Department I; Ludwig-Maximilians-Universität (LMU) Munich; Pettenkoferstrasse 11 D-80336 Munich Germany
| |
Collapse
|
30
|
Spindler V, Waschke J. Desmosomal Cadherins and Signaling: Lessons from Autoimmune Disease. ACTA ACUST UNITED AC 2014; 21:77-84. [DOI: 10.3109/15419061.2013.877000] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
31
|
Abstract
Desmosomes anchor intermediate filaments at sites of cell contact established by the interaction of cadherins extending from opposing cells. The incorporation of cadherins, catenin adaptors, and cytoskeletal elements resembles the closely related adherens junction. However, the recruitment of intermediate filaments distinguishes desmosomes and imparts a unique function. By linking the load-bearing intermediate filaments of neighboring cells, desmosomes create mechanically contiguous cell sheets and, in so doing, confer structural integrity to the tissues they populate. This trait and a well-established role in human disease have long captured the attention of cell biologists, as evidenced by a publication record dating back to the mid-1860s. Likewise, emerging data implicating the desmosome in signaling events pertinent to organismal development, carcinogenesis, and genetic disorders will secure a prominent role for desmosomes in future biological and biomedical investigations.
Collapse
Affiliation(s)
- Robert M Harmon
- Department of Pathology, Northwestern University Feinberg, School of Medicine , Chicago, IL , USA
| | | |
Collapse
|
32
|
Al-Jassar C, Bikker H, Overduin M, Chidgey M. Mechanistic basis of desmosome-targeted diseases. J Mol Biol 2013; 425:4006-22. [PMID: 23911551 PMCID: PMC3807649 DOI: 10.1016/j.jmb.2013.07.035] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 07/23/2013] [Accepted: 07/24/2013] [Indexed: 11/21/2022]
Abstract
Desmosomes are dynamic junctions between cells that maintain the structural integrity of skin and heart tissues by withstanding shear forces. Mutations in component genes cause life-threatening conditions including arrhythmogenic right ventricular cardiomyopathy, and desmosomal proteins are targeted by pathogenic autoantibodies in skin blistering diseases such as pemphigus. Here, we review a set of newly discovered pathogenic alterations and discuss the structural repercussions of debilitating mutations on desmosomal proteins. The architectures of native desmosomal assemblies have been visualized by cryo-electron microscopy and cryo-electron tomography, and the network of protein domain interactions is becoming apparent. Plakophilin and desmoplakin mutations have been discovered to alter binding interfaces, structures, and stabilities of folded domains that have been resolved by X-ray crystallography and NMR spectroscopy. The flexibility within desmoplakin has been revealed by small-angle X-ray scattering and fluorescence assays, explaining how mechanical stresses are accommodated. These studies have shown that the structural and functional consequences of desmosomal mutations can now begin to be understood at multiple levels of spatial and temporal resolution. This review discusses the recent structural insights and raises the possibility of using modeling for mechanism-based diagnosis of how deleterious mutations alter the integrity of solid tissues.
Collapse
Affiliation(s)
- Caezar Al-Jassar
- School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | | | | | | |
Collapse
|
33
|
Choi IK, Strauss R, Richter M, Yun CO, Lieber A. Strategies to increase drug penetration in solid tumors. Front Oncol 2013; 3:193. [PMID: 23898462 PMCID: PMC3724174 DOI: 10.3389/fonc.2013.00193] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 07/11/2013] [Indexed: 12/31/2022] Open
Abstract
Despite significant improvement in modalities for treatment of cancer that led to a longer survival period, the death rate of patients with solid tumors has not changed during the last decades. Emerging studies have identified several physical barriers that limit the therapeutic efficacy of cancer therapeutic agents such as monoclonal antibodies, chemotherapeutic agents, anti-tumor immune cells, and gene therapeutics. Most solid tumors are of epithelial origin and, although malignant cells are de-differentiated, they maintain intercellular junctions, a key feature of epithelial cells, both in the primary tumor as well as in metastatic lesions. Furthermore, nests of malignant epithelial tumor cells are shielded by layers of extracellular matrix (ECM) proteins (e.g., collagen, elastin, fibronectin, laminin) whereby tumor vasculature rarely penetrates into the tumor nests. In this chapter, we will review potential strategies to modulate the ECM and epithelial junctions to enhance the intratumoral diffusion and/or to remove physical masking of target receptors on malignant cells. We will focus on peptides that bind to the junction protein desmoglein 2 and trigger intracellular signaling, resulting in the transient opening of intercellular junctions. Intravenous injection of these junction openers increased the efficacy and safety of therapies with monoclonal antibodies, chemotherapeutics, and T cells in mouse tumor models and was safe in non-human primates. Furthermore, we will summarize approaches to transiently degrade ECM proteins or downregulate their expression. Among these approaches is the intratumoral expression of relaxin or decorin after adenovirus- or stem cell-mediated gene transfer. We will provide examples that relaxin-based approaches increase the anti-tumor efficacy of oncolytic viruses, monoclonal antibodies, and T cells.
Collapse
Affiliation(s)
- Il-Kyu Choi
- Department of Bioengineering, College of Engineering, Hanyang University , Seoul , South Korea
| | | | | | | | | |
Collapse
|
34
|
Nekrasova O, Green KJ. Desmosome assembly and dynamics. Trends Cell Biol 2013; 23:537-46. [PMID: 23891292 DOI: 10.1016/j.tcb.2013.06.004] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 06/14/2013] [Accepted: 06/17/2013] [Indexed: 01/06/2023]
Abstract
Desmosomes are intercellular junctions that anchor intermediate filaments (IFs) to the plasma membrane, forming a supracellular scaffold that provides mechanical resilience to tissues. This anchoring function is accomplished by specialized members of the cadherin family and associated cytoskeletal linking proteins, which together form a highly organized membrane core flanked by mirror-image cytoplasmic plaques. Due to the biochemical insolubility of desmosomes, the mechanisms that govern assembly of these components into a functional organelle remained elusive. Recently developed molecular reporters and live cell imaging approaches have provided powerful new tools to monitor this finely tuned process in real time. Here we discuss studies that are beginning to decipher the machinery and regulation governing desmosome assembly and homeostasis in situ and how these mechanisms are affected during disease pathogenesis.
Collapse
Affiliation(s)
- Oxana Nekrasova
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | |
Collapse
|
35
|
Harmon RM, Simpson CL, Johnson JL, Koetsier JL, Dubash AD, Najor NA, Sarig O, Sprecher E, Green KJ. Desmoglein-1/Erbin interaction suppresses ERK activation to support epidermal differentiation. J Clin Invest 2013; 123:1556-70. [PMID: 23524970 DOI: 10.1172/jci65220] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 01/17/2013] [Indexed: 01/27/2023] Open
Abstract
Genetic disorders of the Ras/MAPK pathway, termed RASopathies, produce numerous abnormalities, including cutaneous keratodermas. The desmosomal cadherin, desmoglein-1 (DSG1), promotes keratinocyte differentiation by attenuating MAPK/ERK signaling and is linked to striate palmoplantar keratoderma (SPPK). This raises the possibility that cutaneous defects associated with SPPK and RASopathies share certain molecular faults. To identify intermediates responsible for executing the inhibition of ERK by DSG1, we conducted a yeast 2-hybrid screen. The screen revealed that Erbin (also known as ERBB2IP), a known ERK regulator, binds DSG1. Erbin silencing disrupted keratinocyte differentiation in culture, mimicking aspects of DSG1 deficiency. Furthermore, ERK inhibition and the induction of differentiation markers by DSG1 required both Erbin and DSG1 domains that participate in binding Erbin. Erbin blocks ERK signaling by interacting with and disrupting Ras-Raf scaffolds mediated by SHOC2, a protein genetically linked to the RASopathy, Noonan-like syndrome with loose anagen hair (NS/LAH). DSG1 overexpression enhanced this inhibitory function, increasing Erbin-SHOC2 interactions and decreasing Ras-SHOC2 interactions. Conversely, analysis of epidermis from DSG1-deficient patients with SPPK demonstrated increased Ras-SHOC2 colocalization and decreased Erbin-SHOC2 colocalization, offering a possible explanation for the observed epidermal defects. These findings suggest a mechanism by which DSG1 and Erbin cooperate to repress MAPK signaling and promote keratinocyte differentiation.
Collapse
Affiliation(s)
- Robert M Harmon
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kowalczyk AP, Green KJ. Structure, function, and regulation of desmosomes. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 116:95-118. [PMID: 23481192 DOI: 10.1016/b978-0-12-394311-8.00005-4] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Desmosomes are adhesive intercellular junctions that mechanically integrate adjacent cells by coupling adhesive interactions mediated by desmosomal cadherins to the intermediate filament cytoskeletal network. Desmosomal cadherins are connected to intermediate filaments by densely clustered cytoplasmic plaque proteins comprising members of the armadillo gene family, including plakoglobin and plakophilins, and members of the plakin family of cytolinkers, such as desmoplakin. The importance of desmosomes in tissue integrity is highlighted by human diseases caused by mutations in desmosomal genes, autoantibody attack of desmosomal cadherins, and bacterial toxins that selectively target desmosomal cadherins. In addition to reviewing the well-known roles of desmosomal proteins in tissue integrity, this chapter also highlights the growing appreciation for how desmosomal proteins are integrated with cell signaling pathways to contribute to vertebrate tissue organization and differentiation.
Collapse
Affiliation(s)
- Andrew P Kowalczyk
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | | |
Collapse
|