1
|
Messingschlager M, Mackowiak SD, Voelker MT, Bieg M, Loske J, Chua RL, Liebig J, Lukassen S, Thürmann L, Seegebarth A, Twardziok S, Doncevic D, Herrmann C, Lorenz S, Klages S, Steinbeis F, Witzenrath M, Kurth F, Conrad C, Sander LE, Ishaque N, Eils R, Lehmann I, Laudi S, Trump S. DNA methylation changes during acute COVID-19 are associated with long-term transcriptional dysregulation in patients' airway epithelial cells. EMBO Mol Med 2025; 17:923-937. [PMID: 40119174 DOI: 10.1038/s44321-025-00215-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/24/2025] Open
Abstract
Molecular changes underlying the persistent health effects after SARS-CoV-2 infection remain poorly understood. To discern the gene regulatory landscape in the upper respiratory tract of COVID-19 patients, we performed enzymatic DNA methylome and single-cell RNA sequencing in nasal cells of COVID-19 patients (n = 19, scRNA-seq n = 14) and controls (n = 14, scRNA-seq n = 10). In addition, we resampled a subset of these patients for transcriptome analyses at 3 (n = 7) and 12 months (n = 5) post infection and followed the expression of differentially regulated genes over time. Genome-wide DNA methylation analysis revealed 3112 differentially methylated regions between COVID-19 patients and controls. Hypomethylated regions affected immune regulatory genes, while hypermethylated regions were associated with genes governing ciliary function. These genes were not only downregulated in the acute phase of the disease but sustained repressed up to 12 months post infection in ciliated cells. Validation in an independent cohort collected 6 months post infection (n = 15) indicated symptom-dependent transcriptional repression of ciliary genes. We therefore propose that hypermethylation observed in the acute phase may exert a long-term effect on gene expression, possibly contributing to post-acute COVID-19 sequelae.
Collapse
Affiliation(s)
- Marey Messingschlager
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Berlin, Germany
- Freie Universität Berlin, Institute of Biology, Berlin, Germany
| | - Sebastian D Mackowiak
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
| | - Maria Theresa Voelker
- Department of Anesthesiology and Intensive Care, University Hospital Leipzig, Leipzig, Germany
| | - Matthias Bieg
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
| | - Jennifer Loske
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Berlin, Germany
- Freie Universität Berlin, Institute of Biology, Berlin, Germany
| | - Robert Lorenz Chua
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
| | - Johannes Liebig
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
| | - Sören Lukassen
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
| | - Loreen Thürmann
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Berlin, Germany
| | - Anke Seegebarth
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Berlin, Germany
| | - Sven Twardziok
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
| | - Daria Doncevic
- Health Data Science Unit, Heidelberg University Hospital and BioQuant, University of Heidelberg, Heidelberg, Germany
| | - Carl Herrmann
- Health Data Science Unit, Heidelberg University Hospital and BioQuant, University of Heidelberg, Heidelberg, Germany
| | - Stephan Lorenz
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Sven Klages
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Fridolin Steinbeis
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany
| | - Martin Witzenrath
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany
- German Center for Lung Research (DZL), Giessen, Germany
| | - Florian Kurth
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Department of Medicine, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Conrad
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
| | - Leif E Sander
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany
- German Center for Lung Research (DZL), Giessen, Germany
| | - Naveed Ishaque
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
| | - Roland Eils
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Berlin, Germany
- Health Data Science Unit, Heidelberg University Hospital and BioQuant, University of Heidelberg, Heidelberg, Germany
- German Center for Lung Research (DZL), Giessen, Germany
- Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin, Germany
| | - Irina Lehmann
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Berlin, Germany
- German Center for Lung Research (DZL), Giessen, Germany
| | - Sven Laudi
- Department of Anesthesiology and Intensive Care, University Hospital Leipzig, Leipzig, Germany
| | - Saskia Trump
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Digital Health, Molecular Epidemiology Unit, Berlin, Germany.
| |
Collapse
|
2
|
Koubova K, Tauber Z, Cizkova K. Exploring the impact of sEH inhibition on intestinal cell differentiation and Colon Cancer: Insights from TPPU treatment. Toxicol Appl Pharmacol 2024; 492:117128. [PMID: 39414156 DOI: 10.1016/j.taap.2024.117128] [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: 07/08/2024] [Revised: 09/20/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
Inhibition of soluble epoxide hydrolase (sEH) appears to be promising for the treatment of many diseases. Studies have focused on the beneficial effects of epoxyeicosatrienoic acids (EETs), which are sEH substrates. However, our recent studies have shown that the sEH activity is crucial for the proper intestinal cell differentiation. In this recent study, we investigated the impact of TPPU, an inhibitor of sEH, on the colon cancer cell lines Caco2 and HT-29. We analysed the changes in the expression of the cytoskeletal protein ezrin and the phosphorylated protein kinase p38 (p-p38). Our results showed a decrease in ezrin expression in differentiated cells and an increase in p-p38 expression after TPPU treatment. Immunocytochemical staining revealed a higher staining intensity of p-p38 in the nuclei of HT-29 cells following TPPU treatment. Immunohistochemical staining was performed on human samples of normal colon tissue, grade 2 tumours, and embryonal/foetal tissues. The staining intensity of ezrin in tumours was reduced in the surface area compared to the crypts. Additionally, we observed the translocation of p-p38 expression from the cytoplasm to the nucleus during differentiation. The tumour samples exhibited higher levels of p-p38 in the cytoplasm, similar to normal undifferentiated tissue. To observe the disruption of the cytoskeleton after TPPU treatment, confocal microscopy was used. It was found that β-actin associated with ezrin forms clusters under the plasma membranes. All of these results are significant because sEH inhibitors are being tested in clinical trials, but they could cause an unexpected adverse effects.
Collapse
Affiliation(s)
- Katerina Koubova
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| | - Zdenek Tauber
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| | - Katerina Cizkova
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic.
| |
Collapse
|
3
|
Yu H, Luo C, Linghu R, Yang J, Wu H. Ezrin Contributes to the Damage of Airway Epithelial Barrier Related to Diabetes Mellitus. J Inflamm Res 2024; 17:2609-2621. [PMID: 38689797 PMCID: PMC11060175 DOI: 10.2147/jir.s449487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 04/18/2024] [Indexed: 05/02/2024] Open
Abstract
Background Diabetes mellitus predisposes individuals to respiratory infections. The airway epithelial barrier provides defense against inhaled antigens and pathogens. Ezrin, is a component of the membrane-cytoskeleton that maintains the cellular morphology, intercellular adhesion, and barrier function of epithelial cells. This study aimed to explore the role of ezrin in airway epithelial barrier damage and correlate its expression and activation with diabetes mellitus. Methods This study was performed in a murine model of diabetes mellitus and with human bronchial epithelial BEAS-2B cells using real-time PCR, Western blotting, immunohistochemical and immunofluorescence staining. Ezrin was knocked down in BEAS-2B cells using siRNA. Ezrin phosphorylation levels were measured to determine activation status. The integrity of the airway epithelial barrier was assessed in vivo by characterizing morphological structure, and in vitro in BEAS-2B cells by measuring tight junction protein expression, transepithelial electrical resistance (TER) and permeability. Results We demonstrated that ezrin expression levels were lower in the lung tissue and airway epithelium of diabetic mice than those in control mice. The morphological structure of the airway epithelium was altered in diabetic mice. High glucose levels downregulated the expression and distribution of ezrin and connexin 43, reduced the expression of tight junction proteins, and altered the epithelial barrier characteristics of BEAS-2B cells. Ezrin knockdown had effects similar to those of high glucose levels. Moreover, a specific inhibitor of ezrin Thr567 phosphorylation (NSC305787) inhibited epithelial barrier formation. Conclusion These results demonstrate that ezrin expression and activation are associated with airway epithelial damage in diabetes mellitus. These findings provide new insights into the molecular pathogenesis of pulmonary infections in diabetes mellitus and may lead to novel therapeutic interventions for airway epithelial barrier damage.
Collapse
Affiliation(s)
- Hongmei Yu
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Cheng Luo
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Ru Linghu
- Department of Internal Medicine, Hospital of Chongqing University, Chongqing, People’s Republic of China
| | - Juan Yang
- Department of Respiratory Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, People’s Republic of China
| | - Haiqiao Wu
- Department of Respiratory Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, People’s Republic of China
| |
Collapse
|
4
|
Kawaguchi K, Yokoyama S, Wang H, Asano S. Inhibition of ezrin phosphorylation by NSC305787 attenuates procaterol-stimulated ciliary beating in airway cilia. Biochem Biophys Res Commun 2024; 703:149685. [PMID: 38373381 DOI: 10.1016/j.bbrc.2024.149685] [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: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/21/2024]
Abstract
Ciliary beating in the airway epithelium plays an important role in preventing infection by eliminating small particles and pathogens. Stimulation of β2 adrenergic receptor (β2AR) increases [cAMP]i levels and strongly activates this ciliary beating. β2AR is localized to the apical membrane of the airways by indirectly binding to ezrin, an actin-binding protein. Ezrin takes active phosphorylated and inactive dephosphorylated states at Thr-567. Previously we showed that procaterol-stimulated ciliary beating was impaired in the ezrin-knockdown mice. In this study, we examined the roles of ezrin and its phosphorylation in regulating ciliary beating by using NSC305787, an ezrin inhibitor, in normal human airway epithelial cells (NHBE). We found that NSC305787 inhibits the phosphorylation of ezrin with an IC50 of 50 μM in NHBE. Treatment with NSC305787 for 4 h or more decreased the expression of β2AR in the cell membrane and induced vesicle- or dot-like expression of ezrin and β2AR inside the cell. As a result, inhibition of ezrin phosphorylation by NSC305787 attenuated the effect of procaterol-induced activation of ciliary beating in both frequency and distance indices.
Collapse
Affiliation(s)
- Kotoku Kawaguchi
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, 525-8577, Japan.
| | - Sakura Yokoyama
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, 525-8577, Japan
| | - Haoting Wang
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, 525-8577, Japan; Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Shinji Asano
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, 525-8577, Japan
| |
Collapse
|
5
|
Kawaguchi K, Tsuji S, Hirao T, Liu Y, Boshi Z, Asano S. Adenosine Stimulates Beating of Neonatal Brain-Derived Cilia through Adenosine A 2B Receptor on the Cilia and Activation of Protein Kinase A Pathway. Biol Pharm Bull 2024; 47:1113-1118. [PMID: 38839362 DOI: 10.1248/bpb.b23-00913] [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: 06/07/2024]
Abstract
Motile cilia in the ependymal cells that line the brain ventricles play pivotal roles in cerebrospinal fluid (CSF) flow in well-defined directions. However, the substances and pathways which regulate their beating have not been well studied. Here, we used primary cultured cells derived from neonatal mouse brain that possess motile cilia and found that adenosine (ADO) stimulates ciliary beating by increasing the ciliary beat frequency (CBF) in a concentration-dependent manner, with the ED50 value being 5 µM. Ciliary beating stimulated by ADO was inhibited by A2B receptor (A2BR) antagonist MRS1754 without any inhibition by antagonists of other ADO receptor subtypes. The expression of A2BR on the cilia was also confirmed by immunofluorescence. The values of CBF were also increased by forskolin, which is an activator of adenylate cyclase, whereas they were not further increased by the addition of ADO. Furthermore, ciliary beating was not stimulated by ADO in the presence of a protein kinase A (PKA) inhibitors. These results altogether suggest that ADO stimulates ciliary beating through A2BR on the cilia, and activation of PKA.
Collapse
Affiliation(s)
- Kotoku Kawaguchi
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University
| | - Suzuka Tsuji
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University
| | - Takuya Hirao
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University
| | - Yixin Liu
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University
| | - Zhao Boshi
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University
| | - Shinji Asano
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University
| |
Collapse
|
6
|
Marunaka Y. Physiological roles of chloride ions in bodily and cellular functions. J Physiol Sci 2023; 73:31. [PMID: 37968609 PMCID: PMC10717538 DOI: 10.1186/s12576-023-00889-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/31/2023] [Indexed: 11/17/2023]
Abstract
Physiological roles of Cl-, a major anion in the body, are not well known compared with those of cations. This review article introduces: (1) roles of Cl- in bodily and cellular functions; (2) the range of cytosolic Cl- concentration ([Cl-]c); (3) whether [Cl-]c could change with cell volume change under an isosmotic condition; (4) whether [Cl-]c could change under conditions where multiple Cl- transporters and channels contribute to Cl- influx and efflux in an isosmotic state; (5) whether the change in [Cl-]c could be large enough to act as signals; (6) effects of Cl- on cytoskeletal tubulin polymerization through inhibition of GTPase activity and tubulin polymerization-dependent biological activity; (7) roles of cytosolic Cl- in cell proliferation; (8) Cl--regulatory mechanisms of ciliary motility; (9) roles of Cl- in sweet/umami taste receptors; (10) Cl--regulatory mechanisms of with-no-lysine kinase (WNK); (11) roles of Cl- in regulation of epithelial Na+ transport; (12) relationship between roles of Cl- and H+ in body functions.
Collapse
Affiliation(s)
- Yoshinori Marunaka
- Medical Research Institute, Kyoto Industrial Health Association, General Incorporated Foundation, 67 Kitatsuboi-Cho, Nishinokyo, Nakagyo-Ku, Kyoto, 604-8472, Japan.
- Research Organization of Science and Technology, Ritsumeikan University, Kusatsu, 525-8577, Japan.
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-Ku, Kyoto, 602-8566, Japan.
| |
Collapse
|
7
|
Dumenil T, Le TT, Rawle DJ, Yan K, Tang B, Nguyen W, Bishop C, Suhrbier A. Warmer ambient air temperatures reduce nasal turbinate and brain infection, but increase lung inflammation in the K18-hACE2 mouse model of COVID-19. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 859:160163. [PMID: 36395835 PMCID: PMC9659553 DOI: 10.1016/j.scitotenv.2022.160163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/04/2022] [Accepted: 11/09/2022] [Indexed: 06/16/2023]
Abstract
Warmer climatic conditions have been associated with fewer COVID-19 cases. Herein we infected K18-hACE2 mice housed at the standard animal house temperature of ∼22 °C, or at ∼31 °C, which is considered to be thermoneutral for mice. On day 2 post infection, RNA-Seq analyses showed no significant differential gene expression lung in lungs of mice housed at the two temperatures, with almost identical viral loads and type I interferon responses. There was also no significant difference in viral loads in lungs on day 5, but RNA-Seq and histology analyses showed clearly elevated inflammatory signatures and infiltrates. Thermoneutrality thus promoted lung inflammation. On day 2 post infection mice housed at 31 °C showed reduced viral loads in nasal turbinates, consistent with increased mucociliary clearance at the warmer ambient temperature. These mice also had reduced virus levels in the brain, and an ensuing amelioration of weight loss and a delay in mortality. Warmer air temperatures may thus reduce infection of the upper respiratory track and the olfactory epithelium, resulting in reduced brain infection. Potential relevance for anosmia and neurological sequelae in COVID-19 patients is discussed.
Collapse
Affiliation(s)
- Troy Dumenil
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4029, Australia
| | - Thuy T Le
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4029, Australia
| | - Daniel J Rawle
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4029, Australia
| | - Kexin Yan
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4029, Australia
| | - Bing Tang
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4029, Australia
| | - Wilson Nguyen
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4029, Australia
| | - Cameron Bishop
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4029, Australia
| | - Andreas Suhrbier
- Immunology Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4029, Australia; Australian Infectious Disease Research Centre, GVN Center of Excellence, Brisbane, Queensland 4029, 4072, Australia.
| |
Collapse
|
8
|
Zhao S, Luo J, Hu J, Wang H, Zhao N, Cao M, Zhang C, Hu R, Liu L. Role of Ezrin in Asthma-Related Airway Inflammation and Remodeling. Mediators Inflamm 2022; 2022:6255012. [PMID: 36530558 PMCID: PMC9750775 DOI: 10.1155/2022/6255012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 08/13/2023] Open
Abstract
Ezrin is an actin binding protein connecting the cell membrane and the cytoskeleton, which is crucial to maintaining cell morphology, intercellular adhesion, and cytoskeleton remodeling. Asthma involves dysfunction of inflammatory cells, cytokines, and airway structural cells. Recent studies have shown that ezrin, whose function is affected by extensive phosphorylation and protein interactions, is closely associated with asthma, may be a therapeutic target for asthma treatment. In this review, we summarize studies on ezrin and discuss its role in asthma-related airway inflammation and remodeling.
Collapse
Affiliation(s)
- Shumei Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Jiaqi Luo
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Jun Hu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Hesheng Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Ningwei Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Shimadzu Biomedical Research Laboratory, Shanghai 200233, China
| | - Meng Cao
- Nanjing University of Chinese Medicine, Nanjing 210029, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Cong Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Rongkui Hu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Lanying Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Nanjing University of Chinese Medicine, Nanjing 210029, China
| |
Collapse
|
9
|
First person – Kotoku Kawaguchi. J Cell Sci 2022. [DOI: 10.1242/jcs.259961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
ABSTRACT
First Person is a series of interviews with the first authors of a selection of papers published in Journal of Cell Science, helping early-career researchers promote themselves alongside their papers. Kotoku Kawaguchi is first author on ‘ Ezrin knockdown reduces procaterol-stimulated ciliary beating without morphological changes in mouse airway cilia’, published in JCS. Kotoku is an assistant professor in the lab of Shinji Asano at Ritsumeikan University, Kusatsu, Japan, investigating the role of ezrin in ciliary beating and mucociliary clearance.
Collapse
|
10
|
Kawaguchi K, Asano S. Pathophysiological Roles of Actin-Binding Scaffold Protein, Ezrin. Int J Mol Sci 2022; 23:ijms23063246. [PMID: 35328667 PMCID: PMC8952289 DOI: 10.3390/ijms23063246] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/14/2022] [Accepted: 03/14/2022] [Indexed: 02/06/2023] Open
Abstract
Ezrin is one of the members of the ezrin/radixin/moesin (ERM) family of proteins. It was originally discovered as an actin-binding protein in the microvilli structure about forty years ago. Since then, it has been revealed as a key protein with functions in a variety of fields including cell migration, survival, and signal transduction, as well as functioning as a structural component. Ezrin acts as a cross-linker of membrane proteins or phospholipids in the plasma membrane and the actin cytoskeleton. It also functions as a platform for signaling molecules at the cell surface. Moreover, ezrin is regarded as an important target protein in cancer diagnosis and therapy because it is a key protein involved in cancer progression and metastasis, and its high expression is linked to poor survival in many cancers. Small molecule inhibitors of ezrin have been developed and investigated as candidate molecules that suppress cancer metastasis. Here, we wish to comprehensively review the roles of ezrin from the pathophysiological points of view.
Collapse
|