1
|
Ran Q, Li A, Tan Y, Zhang Y, Zhang Y, Chen H. Action and therapeutic targets of myosin light chain kinase, an important cardiovascular signaling mechanism. Pharmacol Res 2024; 206:107276. [PMID: 38944220 DOI: 10.1016/j.phrs.2024.107276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 06/19/2024] [Indexed: 07/01/2024]
Abstract
The global incidence of cardiac diseases is increasing, imposing a substantial socioeconomic burden on healthcare systems. The pathogenesis of cardiovascular disease is complex and not fully understood, and the physiological function of the heart is inextricably linked to well-regulated cardiac muscle movement. Myosin light chain kinase (MLCK) is essential for myocardial contraction and diastole, cardiac electrophysiological homeostasis, vasoconstriction of vascular nerves and blood pressure regulation. In this sense, MLCK appears to be an attractive therapeutic target for cardiac diseases. MLCK participates in myocardial cell movement and migration through diverse pathways, including regulation of calcium homeostasis, activation of myosin light chain phosphorylation, and stimulation of vascular smooth muscle cell contraction or relaxation. Recently, phosphorylation of myosin light chains has been shown to be closely associated with the activation of myocardial exercise signaling, and MLCK mediates systolic and diastolic functions of the heart through the interaction of myosin thick filaments and actin thin filaments. It works by upholding the integrity of the cytoskeleton, modifying the conformation of the myosin head, and modulating innervation. MLCK governs vasoconstriction and diastolic function and is associated with the activation of adrenergic and sympathetic nervous systems, extracellular transport, endothelial permeability, and the regulation of nitric oxide and angiotensin II. Additionally, MLCK plays a crucial role in the process of cardiac aging. Multiple natural products/phytochemicals and chemical compounds, such as quercetin, cyclosporin, and ML-7 hydrochloride, have been shown to regulate cardiomyocyte MLCK. The MLCK-modifying capacity of these compounds should be considered in designing novel therapeutic agents. This review summarizes the mechanism of action of MLCK in the cardiovascular system and the therapeutic potential of reported chemical compounds in cardiac diseases by modifying MLCK processes.
Collapse
Affiliation(s)
- Qingzhi Ran
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing 100070, China
| | - Aoshuang Li
- Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing 100053, China
| | - Yuqing Tan
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing 100070, China
| | - Yue Zhang
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing 100070, China.
| | - Yongkang Zhang
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China.
| | - Hengwen Chen
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing 100070, China.
| |
Collapse
|
2
|
Chanez-Paredes SD, Abtahi S, Zha J, Li E, Marsischky G, Zuo L, Grey MJ, He W, Turner JR. Mechanisms underlying distinct subcellular localization and regulation of epithelial long myosin light-chain kinase splice variants. J Biol Chem 2024; 300:105643. [PMID: 38199574 PMCID: PMC10862019 DOI: 10.1016/j.jbc.2024.105643] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 12/13/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Intestinal epithelia express two long myosin light-chain kinase (MLCK) splice variants, MLCK1 and MLCK2, which differ by the absence of a complete immunoglobulin (Ig)-like domain 3 within MLCK2. MLCK1 is preferentially associated with the perijunctional actomyosin ring at steady state, and this localization is enhanced by inflammatory stimuli including tumor necrosis factor (TNF). Here, we sought to identify MLCK1 domains that direct perijunctional MLCK1 localization and their relevance to disease. Ileal biopsies from Crohn's disease patients demonstrated preferential increases in MLCK1 expression and perijunctional localization relative to healthy controls. In contrast to MLCK1, MLCK2 expressed in intestinal epithelia is predominantly associated with basal stress fibers, and the two isoforms have distinct effects on epithelial migration and barrier regulation. MLCK1(Ig1-4) and MLCK1(Ig1-3), but not MLCK2(Ig1-4) or MLCK1(Ig3), directly bind to F-actin in vitro and direct perijunctional recruitment in intestinal epithelial cells. Further study showed that Ig1 is unnecessary, but that, like Ig3, the unstructured linker between Ig1 and Ig2 (Ig1/2us) is essential for recruitment. Despite being unable to bind F-actin or direct recruitment independently, Ig3 does have dominant negative functions that allow it to displace perijunctional MLCK1, increase steady-state barrier function, prevent TNF-induced MLCK1 recruitment, and attenuate TNF-induced barrier loss. These data define the minimal domain required for MLCK1 localization and provide mechanistic insight into the MLCK1 recruitment process. Overall, the results create a foundation for development of molecularly targeted therapies that target key domains to prevent MLCK1 recruitment, restore barrier function, and limit inflammatory bowel disease progression.
Collapse
Affiliation(s)
- Sandra D Chanez-Paredes
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Shabnam Abtahi
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Juanmin Zha
- Department of Oncology, The First Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genomic Resource Center, Suzhou Medical School of Soochow University, Suzhou, China
| | - Enkai Li
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gerald Marsischky
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Li Zuo
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA; Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui, China
| | - Michael J Grey
- Gastroenterology Division, Department of Medicine, Beth-Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Weiqi He
- Department of Oncology, The First Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genomic Resource Center, Suzhou Medical School of Soochow University, Suzhou, China.
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
3
|
Rossi F, Liu M, Tieniber A, Etherington MS, Hanna A, Vitiello GA, Param NJ, Do K, Wang L, Antonescu CR, Zeng S, Zhang JQ, DeMatteo RP. Myosin Light-Chain Kinase Inhibition Potentiates the Antitumor Effects of Avapritinib in PDGFRA D842V-Mutant Gastrointestinal Stromal Tumor. Clin Cancer Res 2023; 29:2144-2157. [PMID: 36971786 PMCID: PMC10239357 DOI: 10.1158/1078-0432.ccr-22-0533] [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: 02/16/2022] [Revised: 01/20/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023]
Abstract
PURPOSE To create an in vivo model of PDGFRA D842V-mutant gastrointestinal stromal tumor (GIST) and identify the mechanism of tumor persistence following avapritinib therapy. EXPERIMENTAL DESIGN We created a patient-derived xenograft (PDX) of PDGFRA D842V-mutant GIST and tested the effects of imatinib, avapritinib, and ML-7, an inhibitor of myosin light-chain kinase (MYLK). Bulk tumor RNA sequencing and oncogenic signaling were evaluated. Apoptosis, survival, and actin cytoskeleton were evaluated in GIST T1 cells and isolated PDX cells in vitro. Human GIST specimens were analyzed for MYLK expression. RESULTS The PDX was minimally responsive to imatinib but sensitive to avapritinib. Avapritinib therapy increased tumor expression of genes related to the actin cytoskeleton, including MYLK. ML-7 induced apoptosis and disrupted actin filaments in short-term cultures of PDX cells and decreased survival in GIST T1 cells in combination with imatinib or avapritinib. Combined therapy with ML-7 improved the antitumor effects of low-dose avapritinib in vivo. Furthermore, MYLK was expressed in human GIST specimens. CONCLUSIONS MYLK upregulation is a novel mechanism of tumor persistence after tyrosine kinase inhibition. Concomitant MYLK inhibition may enable the use of a lower dose of avapritinib, which is associated with dose-dependent cognitive side effects.
Collapse
Affiliation(s)
- Ferdinand Rossi
- Perelman School of Medicine, Department of Surgery, University of Pennsylvania, Philadelphia PA
| | - Mengyuan Liu
- Perelman School of Medicine, Department of Surgery, University of Pennsylvania, Philadelphia PA
| | - Andrew Tieniber
- Perelman School of Medicine, Department of Surgery, University of Pennsylvania, Philadelphia PA
| | - Mark S. Etherington
- Perelman School of Medicine, Department of Surgery, University of Pennsylvania, Philadelphia PA
| | - Andrew Hanna
- Perelman School of Medicine, Department of Surgery, University of Pennsylvania, Philadelphia PA
| | - Gerardo A. Vitiello
- Perelman School of Medicine, Department of Surgery, University of Pennsylvania, Philadelphia PA
| | - Nesteene J. Param
- Perelman School of Medicine, Department of Surgery, University of Pennsylvania, Philadelphia PA
| | - Kevin Do
- Perelman School of Medicine, Department of Surgery, University of Pennsylvania, Philadelphia PA
| | - Laura Wang
- Perelman School of Medicine, Department of Surgery, University of Pennsylvania, Philadelphia PA
| | | | - Shan Zeng
- Perelman School of Medicine, Department of Surgery, University of Pennsylvania, Philadelphia PA
| | - Jennifer Q. Zhang
- Perelman School of Medicine, Department of Surgery, University of Pennsylvania, Philadelphia PA
| | - Ronald P. DeMatteo
- Perelman School of Medicine, Department of Surgery, University of Pennsylvania, Philadelphia PA
| |
Collapse
|
4
|
Berger SL, Leo-Macias A, Yuen S, Khatri L, Pfennig S, Zhang Y, Agullo-Pascual E, Caillol G, Zhu MS, Rothenberg E, Melendez-Vasquez CV, Delmar M, Leterrier C, Salzer JL. Localized Myosin II Activity Regulates Assembly and Plasticity of the Axon Initial Segment. Neuron 2018; 97:555-570.e6. [PMID: 29395909 PMCID: PMC5805619 DOI: 10.1016/j.neuron.2017.12.039] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 08/24/2017] [Accepted: 12/22/2017] [Indexed: 01/08/2023]
Abstract
The axon initial segment (AIS) is the site of action potential generation and a locus of activity-dependent homeostatic plasticity. A multimeric complex of sodium channels, linked via a cytoskeletal scaffold of ankyrin G and beta IV spectrin to submembranous actin rings, mediates these functions. The mechanisms that specify the AIS complex to the proximal axon and underlie its plasticity remain poorly understood. Here we show phosphorylated myosin light chain (pMLC), an activator of contractile myosin II, is highly enriched in the assembling and mature AIS, where it associates with actin rings. MLC phosphorylation and myosin II contractile activity are required for AIS assembly, and they regulate the distribution of AIS components along the axon. pMLC is rapidly lost during depolarization, destabilizing actin and thereby providing a mechanism for activity-dependent structural plasticity of the AIS. Together, these results identify pMLC/myosin II activity as a common link between AIS assembly and plasticity.
Collapse
Affiliation(s)
- Stephen L Berger
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA
| | | | - Stephanie Yuen
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA
| | - Latika Khatri
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA
| | - Sylvia Pfennig
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA
| | - Yanqing Zhang
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA
| | | | - Ghislaine Caillol
- Aix Marseille Université, CNRS, INP UMR7051, 13344 Cedex 15, Marseille, France
| | - Min-Sheng Zhu
- Model Animal Research Center and MOE Key Laboratory of Model Animal and Disease Study, Nanjing University, Nanjing 210061, China
| | - Eli Rothenberg
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, NY 10016, USA
| | - Carmen V Melendez-Vasquez
- Department of Biological Sciences, Hunter College, New York, NY 10065, USA; Department of Molecular, Cellular, and Developmental Biology, The Graduate Center, The City University of New York, NY 10016, USA
| | - Mario Delmar
- Division of Cardiology, NYU School of Medicine, New York, NY 10016, USA
| | | | - James L Salzer
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
5
|
Khapchaev AY, Shirinsky VP. Myosin Light Chain Kinase MYLK1: Anatomy, Interactions, Functions, and Regulation. BIOCHEMISTRY (MOSCOW) 2017; 81:1676-1697. [PMID: 28260490 DOI: 10.1134/s000629791613006x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
This review discusses and summarizes the results of molecular and cellular investigations of myosin light chain kinase (MLCK, MYLK1), the key regulator of cell motility. The structure and regulation of a complex mylk1 gene and the domain organization of its products is presented. The interactions of the mylk1 gene protein products with other proteins and posttranslational modifications of the mylk1 gene protein products are reviewed, which altogether might determine the role and place of MLCK in physiological and pathological reactions of cells and entire organisms. Translational potential of MLCK as a drug target is evaluated.
Collapse
Affiliation(s)
- A Y Khapchaev
- Russian Cardiology Research and Production Center, Moscow, 121552, Russia.
| | | |
Collapse
|
6
|
Vilitkevich EL, Khapchaev AY, Kudryashov DS, Nikashin AV, Schavocky JP, Lukas TJ, Watterson DM, Shirinsky VP. Phosphorylation Regulates Interaction of 210-kDa Myosin Light Chain Kinase N-terminal Domain with Actin Cytoskeleton. BIOCHEMISTRY (MOSCOW) 2016; 80:1288-97. [PMID: 26567572 DOI: 10.1134/s0006297915100090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
High molecular weight myosin light chain kinase (MLCK210) is a multifunctional protein involved in myosin II activation and integration of cytoskeletal components in cells. MLCK210 possesses actin-binding regions both in the central part of the molecule and in its N-terminal tail domain. In HeLa cells, mitotic protein kinase Aurora B was suggested to phosphorylate MLCK210 N-terminal tail at serine residues (Dulyaninova, N. G., and Bresnick, A. R. (2004) Exp. Cell Res., 299, 303-314), but the functional significance of the phosphorylation was not established. We report here that in vitro, the N-terminal actin-binding domain of MLCK210 is located within residues 27-157 (N27-157, avian MLCK210 sequence) and is phosphorylated by cAMP-dependent protein kinase (PKA) and Aurora B at serine residues 140/149 leading to a decrease in N27-157 binding to actin. The same residues are phosphorylated in a PKA-dependent manner in transfected HeLa cells. Further, in transfected cells, phosphomimetic mutants of N27-157 showed reduced association with the detergent-stable cytoskeleton, whereas in vitro, the single S149D mutation reduced N27-157 association with F-actin to a similar extent as that achieved by N27-157 phosphorylation. Altogether, our results indicate that phosphorylation of MLCK210 at distinct serine residues, mainly at S149, attenuates the interaction of MLCK210 N-terminus with the actin cytoskeleton and might serve to regulate MLCK210 microfilament cross-linking activity in cells.
Collapse
Affiliation(s)
- E L Vilitkevich
- Russian Cardiology Research and Production Center, Moscow, 121552, Russia.
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Sundararajan V, Gengenbacher N, Stemmler MP, Kleemann JA, Brabletz T, Brabletz S. The ZEB1/miR-200c feedback loop regulates invasion via actin interacting proteins MYLK and TKS5. Oncotarget 2016; 6:27083-96. [PMID: 26334100 PMCID: PMC4694975 DOI: 10.18632/oncotarget.4807] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 08/07/2015] [Indexed: 02/06/2023] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a developmental process which is aberrantly activated during cancer invasion and metastasis. Elevated expression of EMT-inducers like ZEB1 enables tumor cells to detach from the primary tumor and invade into the surrounding tissue. The main antagonist of ZEB1 in controlling EMT is the microRNA-200 family that is reciprocally linked to ZEB1 in a double negative feedback loop. Here, we further elucidate how the ZEB1/miR-200 feedback loop controls invasion of tumor cells. The process of EMT is attended by major changes in the actin cytoskeleton. Via in silico screening of genes encoding for actin interacting proteins, we identified two novel targets of miR-200c - TKS5 and MYLK (MLCK). Co-expression of both genes with ZEB1 was observed in several cancer cell lines as well as in breast cancer patients and correlated with low miR-200c levels. Depletion of TKS5 or MYLK in breast cancer cells reduced their invasive potential and their ability to form invadopodia. Whereas TKS5 is known to be a major component, we could identify MYLK as a novel player in invadopodia formation. In summary, TKS5 and MYLK represent two mediators of invasive behavior of cancer cells that are regulated by the ZEB1/miR-200 feedback loop.
Collapse
Affiliation(s)
- Vignesh Sundararajan
- Department of Visceral Surgery, University Medical Center Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, Freiburg, Germany.,Faculty of Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Nicolas Gengenbacher
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Marc P Stemmler
- Department of Experimental Medicine I, Nikolaus-Fiebiger-Center for Molecular Medicine, University Erlangen-Nürnberg, Erlangen, Germany
| | - Julia A Kleemann
- Department of Experimental Medicine I, Nikolaus-Fiebiger-Center for Molecular Medicine, University Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine I, Nikolaus-Fiebiger-Center for Molecular Medicine, University Erlangen-Nürnberg, Erlangen, Germany
| | - Simone Brabletz
- Department of Experimental Medicine I, Nikolaus-Fiebiger-Center for Molecular Medicine, University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
8
|
Chen C, Tao T, Wen C, He WQ, Qiao YN, Gao YQ, Chen X, Wang P, Chen CP, Zhao W, Chen HQ, Ye AP, Peng YJ, Zhu MS. Myosin light chain kinase (MLCK) regulates cell migration in a myosin regulatory light chain phosphorylation-independent mechanism. J Biol Chem 2014; 289:28478-88. [PMID: 25122766 DOI: 10.1074/jbc.m114.567446] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Myosin light chain kinase (MLCK) has long been implicated in the myosin phosphorylation and force generation required for cell migration. Here, we surprisingly found that the deletion of MLCK resulted in fast cell migration, enhanced protrusion formation, and no alteration of myosin light chain phosphorylation. The mutant cells showed reduced membrane tether force and fewer membrane F-actin filaments. This phenotype was rescued by either kinase-dead MLCK or five-DFRXXL motif, a MLCK fragment with potent F-actin-binding activity. Pull-down and co-immunoprecipitation assays showed that the absence of MLCK led to attenuated formation of transmembrane complexes, including myosin II, integrins and fibronectin. We suggest that MLCK is not required for myosin phosphorylation in a migrating cell. A critical role of MLCK in cell migration involves regulating the cell membrane tension and protrusion necessary for migration, thereby stabilizing the membrane skeleton through F-actin-binding activity. This finding sheds light on a novel regulatory mechanism of protrusion during cell migration.
Collapse
Affiliation(s)
- Chen Chen
- From the Model Animal Research Center, Key Laboratory of Model Animal for Disease Study of Ministry of Education, Nanjing University, Nanjing 210061, P.R. China
| | - Tao Tao
- From the Model Animal Research Center, Key Laboratory of Model Animal for Disease Study of Ministry of Education, Nanjing University, Nanjing 210061, P.R. China
| | - Cheng Wen
- School of Electronics Engineering and Computer Science, Key Laboratory for the Physics & Chemistry of Nanodevices of Ministry of Education, Peking University, Beijing 100871, P.R. China, and
| | - Wei-Qi He
- From the Model Animal Research Center, Key Laboratory of Model Animal for Disease Study of Ministry of Education, Nanjing University, Nanjing 210061, P.R. China
| | - Yan-Ning Qiao
- From the Model Animal Research Center, Key Laboratory of Model Animal for Disease Study of Ministry of Education, Nanjing University, Nanjing 210061, P.R. China
| | - Yun-Qian Gao
- From the Model Animal Research Center, Key Laboratory of Model Animal for Disease Study of Ministry of Education, Nanjing University, Nanjing 210061, P.R. China
| | - Xin Chen
- From the Model Animal Research Center, Key Laboratory of Model Animal for Disease Study of Ministry of Education, Nanjing University, Nanjing 210061, P.R. China
| | - Pei Wang
- From the Model Animal Research Center, Key Laboratory of Model Animal for Disease Study of Ministry of Education, Nanjing University, Nanjing 210061, P.R. China
| | - Cai-Ping Chen
- From the Model Animal Research Center, Key Laboratory of Model Animal for Disease Study of Ministry of Education, Nanjing University, Nanjing 210061, P.R. China
| | - Wei Zhao
- From the Model Animal Research Center, Key Laboratory of Model Animal for Disease Study of Ministry of Education, Nanjing University, Nanjing 210061, P.R. China
| | - Hua-Qun Chen
- School of Life Science, Nanjing Normal University, Nanjing 210009, P.R. China
| | - An-Pei Ye
- School of Electronics Engineering and Computer Science, Key Laboratory for the Physics & Chemistry of Nanodevices of Ministry of Education, Peking University, Beijing 100871, P.R. China, and
| | - Ya-Jing Peng
- From the Model Animal Research Center, Key Laboratory of Model Animal for Disease Study of Ministry of Education, Nanjing University, Nanjing 210061, P.R. China,
| | - Min-Sheng Zhu
- From the Model Animal Research Center, Key Laboratory of Model Animal for Disease Study of Ministry of Education, Nanjing University, Nanjing 210061, P.R. China, School of Life Science, Nanjing Normal University, Nanjing 210009, P.R. China
| |
Collapse
|
9
|
Wilson JLL, Miranda CA, Knepper MA. Vasopressin and the regulation of aquaporin-2. Clin Exp Nephrol 2013; 17:751-64. [PMID: 23584881 PMCID: PMC3775849 DOI: 10.1007/s10157-013-0789-5] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 02/25/2013] [Indexed: 12/26/2022]
Abstract
Water excretion is regulated in large part through the regulation of osmotic water permeability of the renal collecting duct epithelium. Water permeability is controlled by vasopressin through regulation of the water channel, aquaporin-2 (AQP2). Two processes contribute: (1) regulation of AQP2 trafficking to the apical plasma membrane; and (2) regulation of the total amount of the AQP2 protein in the cells. Regulation of AQP2 abundance is defective in several water-balance disorders, including many polyuric disorders and the syndrome of inappropriate antidiuresis. Here we review vasopressin signaling in the renal collecting duct that is relevant to the two modes of water permeability regulation.
Collapse
Affiliation(s)
- Justin L L Wilson
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 10 Center Dr., Bldg 10, Room 6N260, Bethesda, MD, 20892-1603, USA
| | | | | |
Collapse
|
10
|
Bolger SJ, Hurtado PAG, Hoffert JD, Saeed F, Pisitkun T, Knepper MA. Quantitative phosphoproteomics in nuclei of vasopressin-sensitive renal collecting duct cells. Am J Physiol Cell Physiol 2012; 303:C1006-20. [PMID: 22992673 DOI: 10.1152/ajpcell.00260.2012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vasopressin regulates transport across the collecting duct epithelium in part via effects on gene transcription. Transcriptional regulation occurs partially via changes in phosphorylation of transcription factors, transcriptional coactivators, and protein kinases in the nucleus. To test whether vasopressin alters the nuclear phosphoproteome of vasopressin-sensitive cultured mouse mpkCCD cells, we used stable isotope labeling and mass spectrometry to quantify thousands of phosphorylation sites in nuclear extracts and nuclear pellet fractions. Measurements were made in the presence and absence of the vasopressin analog dDAVP. Of the 1,251 sites quantified, 39 changed significantly in response to dDAVP. Network analysis of the regulated proteins revealed two major clusters ("cell-cell adhesion" and "transcriptional regulation") that were connected to known elements of the vasopressin signaling pathway. The hub proteins for these two clusters were the transcriptional coactivator β-catenin and the transcription factor c-Jun. Phosphorylation of β-catenin at Ser552 was increased by dDAVP [log(2)(dDAVP/vehicle) = 1.79], and phosphorylation of c-Jun at Ser73 was decreased [log(2)(dDAVP/vehicle) = -0.53]. The β-catenin site is known to be targeted by either protein kinase A or Akt, both of which are activated in response to vasopressin. The c-Jun site is a canonical target for the MAP kinase Jnk2, which is downregulated in response to vasopressin in the collecting duct. The data support the idea that vasopressin-mediated control of transcription in collecting duct cells involves selective changes in the nuclear phosphoproteome. All data are available to users at http://helixweb.nih.gov/ESBL/Database/mNPPD/.
Collapse
Affiliation(s)
- Steven J Bolger
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1603, USA
| | | | | | | | | | | |
Collapse
|
11
|
Roca-Cusachs P, Iskratsch T, Sheetz MP. Finding the weakest link: exploring integrin-mediated mechanical molecular pathways. J Cell Sci 2012; 125:3025-38. [PMID: 22797926 DOI: 10.1242/jcs.095794] [Citation(s) in RCA: 184] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
From the extracellular matrix to the cytoskeleton, a network of molecular links connects cells to their environment. Molecules in this network transmit and detect mechanical forces, which subsequently determine cell behavior and fate. Here, we reconstruct the mechanical pathway followed by these forces. From matrix proteins to actin through integrins and adaptor proteins, we review how forces affect the lifetime of bonds and stretch or alter the conformation of proteins, and how these mechanical changes are converted into biochemical signals in mechanotransduction events. We evaluate which of the proteins in the network can participate in mechanotransduction and which are simply responsible for transmitting forces in a dynamic network. Besides their individual properties, we also analyze how the mechanical responses of a protein are determined by their serial connections from the matrix to actin, their parallel connections in integrin clusters and by the rate at which force is applied to them. All these define mechanical molecular pathways in cells, which are emerging as key regulators of cell function alongside better studied biochemical pathways.
Collapse
Affiliation(s)
- Pere Roca-Cusachs
- University of Barcelona and Institute for Bioengineering of Catalonia, Barcelona, Spain.
| | | | | |
Collapse
|
12
|
Zhu GJ, Wang F, Chen C, Xu L, Zhang WC, Fan C, Peng YJ, Chen J, He WQ, Guo SY, Zuo J, Gao X, Zhu MS. Myosin light-chain kinase is necessary for membrane homeostasis in cochlear inner hair cells. PLoS One 2012; 7:e34894. [PMID: 22485190 PMCID: PMC3317649 DOI: 10.1371/journal.pone.0034894] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 03/08/2012] [Indexed: 12/04/2022] Open
Abstract
The structural homeostasis of the cochlear hair cell membrane is critical for all aspects of sensory transduction, but the regulation of its maintenance is not well understood. In this report, we analyzed the cochlear hair cells of mice with specific deletion of myosin light chain kinase (MLCK) in inner hair cells. MLCK-deficient mice showed impaired hearing, with a 5- to 14-dB rise in the auditory brainstem response (ABR) thresholds to clicks and tones of different frequencies and a significant decrease in the amplitude of the ABR waves. The mutant inner hair cells produced several ball-like structures around the hair bundles in vivo, indicating impaired membrane stability. Inner hair cells isolated from the knockout mice consistently displayed less resistance to hypoosmotic solution and less membrane F-actin. Myosin light-chain phosphorylation was also reduced in the mutated inner hair cells. Our results suggest that MLCK is necessary for maintaining the membrane stability of inner hair cells.
Collapse
MESH Headings
- Actin Cytoskeleton/metabolism
- Actins/metabolism
- Animals
- Cell Membrane/enzymology
- Cell Membrane/metabolism
- Epithelium/enzymology
- Epithelium/metabolism
- Evoked Potentials, Auditory, Brain Stem
- Gene Expression
- Hair Cells, Auditory, Inner/enzymology
- Hair Cells, Auditory, Inner/metabolism
- Hair Cells, Auditory, Inner/ultrastructure
- Homeostasis
- Mice
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Myosin Light Chains/metabolism
- Myosin VIIa
- Myosin-Light-Chain Kinase/deficiency
- Myosin-Light-Chain Kinase/genetics
- Myosin-Light-Chain Kinase/physiology
- Myosins/metabolism
- Organ of Corti/cytology
- Osmotic Pressure
- Phosphorylation
- Protein Processing, Post-Translational
- Sequence Deletion
- Sodium-Potassium-Exchanging ATPase/genetics
- Sodium-Potassium-Exchanging ATPase/metabolism
Collapse
Affiliation(s)
- Guang-Jie Zhu
- MOE Key Laboratory for Model Animal and Diseases Studies, Nanjing Drum Tower Hospital and Model Animal Research Center of Nanjing University, Nanjing, China
| | - Fang Wang
- MOE Key Laboratory for Model Animal and Diseases Studies, Nanjing Drum Tower Hospital and Model Animal Research Center of Nanjing University, Nanjing, China
| | - Chen Chen
- MOE Key Laboratory for Model Animal and Diseases Studies, Nanjing Drum Tower Hospital and Model Animal Research Center of Nanjing University, Nanjing, China
| | - Lin Xu
- MOE Key Laboratory for Model Animal and Diseases Studies, Nanjing Drum Tower Hospital and Model Animal Research Center of Nanjing University, Nanjing, China
| | - Wen-Cheng Zhang
- MOE Key Laboratory for Model Animal and Diseases Studies, Nanjing Drum Tower Hospital and Model Animal Research Center of Nanjing University, Nanjing, China
- Zhejiang Provincial Key Lab for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical College, University Park, Wenzhou, China
| | - Chi Fan
- MOE Key Laboratory for Model Animal and Diseases Studies, Nanjing Drum Tower Hospital and Model Animal Research Center of Nanjing University, Nanjing, China
| | - Ya-Jing Peng
- MOE Key Laboratory for Model Animal and Diseases Studies, Nanjing Drum Tower Hospital and Model Animal Research Center of Nanjing University, Nanjing, China
| | - Jie Chen
- MOE Key Laboratory for Model Animal and Diseases Studies, Nanjing Drum Tower Hospital and Model Animal Research Center of Nanjing University, Nanjing, China
| | - Wei-Qi He
- MOE Key Laboratory for Model Animal and Diseases Studies, Nanjing Drum Tower Hospital and Model Animal Research Center of Nanjing University, Nanjing, China
| | - Shi-Ying Guo
- MOE Key Laboratory for Model Animal and Diseases Studies, Nanjing Drum Tower Hospital and Model Animal Research Center of Nanjing University, Nanjing, China
| | - Jian Zuo
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Xia Gao
- MOE Key Laboratory for Model Animal and Diseases Studies, Nanjing Drum Tower Hospital and Model Animal Research Center of Nanjing University, Nanjing, China
- * E-mail: (XG); (M-SZ)
| | - Min-Sheng Zhu
- MOE Key Laboratory for Model Animal and Diseases Studies, Nanjing Drum Tower Hospital and Model Animal Research Center of Nanjing University, Nanjing, China
- Zhejiang Provincial Key Lab for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical College, University Park, Wenzhou, China
- * E-mail: (XG); (M-SZ)
| |
Collapse
|
13
|
Bhuwania R, Cornfine S, Fang Z, Krüger M, Luna EJ, Linder S. Supervillin couples myosin-dependent contractility to podosomes and enables their turnover. J Cell Sci 2012; 125:2300-14. [PMID: 22344260 DOI: 10.1242/jcs.100032] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Podosomes are actin-rich adhesion and invasion structures. Especially in macrophages, podosomes exist in two subpopulations, large precursors at the cell periphery and smaller podosomes (successors) in the cell interior. To date, the mechanisms that differentially regulate these subpopulations are largely unknown. Here, we show that the membrane-associated protein supervillin localizes preferentially to successor podosomes and becomes enriched at precursors immediately before their dissolution. Consistently, podosome numbers are inversely correlated with supervillin protein levels. Using deletion constructs, we find that the myosin II regulatory N-terminus of supervillin [SV(1-174)] is crucial for these effects. Phosphorylated myosin light chain (pMLC) localizes at supervillin-positive podosomes, and time-lapse analyses show that enrichment of GFP-supervillin at podosomes coincides with their coupling to contractile myosin-IIA-positive cables. We also show that supervillin binds only to activated myosin IIA, and a dysregulated N-terminal construct [SV(1-830)] enhances pMLC levels at podosomes. Thus, preferential recruitment of supervillin to podosome subpopulations might both require and induce actomyosin contractility. Using siRNA and pharmacological inhibition, we demonstrate that supervillin and myosin IIA cooperate to regulate podosome lifetime, podosomal matrix degradation and cell polarization. In sum, we show here that podosome subpopulations differ in their molecular composition and identify supervillin, in cooperation with myosin IIA, as a crucial factor in the regulation of podosome turnover and function.
Collapse
Affiliation(s)
- Ridhirama Bhuwania
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Eppendorf, Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
14
|
Molluscan catch muscle myorod and its N-terminal peptide bind to F-actin and myosin in a phosphorylation-dependent manner. Arch Biochem Biophys 2011; 509:59-65. [DOI: 10.1016/j.abb.2011.02.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 02/12/2011] [Accepted: 02/12/2011] [Indexed: 12/18/2022]
|
15
|
Gautel M. Cytoskeletal protein kinases: titin and its relations in mechanosensing. Pflugers Arch 2011; 462:119-34. [PMID: 21416260 PMCID: PMC3114093 DOI: 10.1007/s00424-011-0946-1] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 02/15/2011] [Accepted: 02/18/2011] [Indexed: 12/22/2022]
Abstract
Titin, the giant elastic ruler protein of striated muscle sarcomeres, contains a catalytic kinase domain related to a family of intrasterically regulated protein kinases. The most extensively studied member of this branch of the human kinome is the Ca2+–calmodulin (CaM)-regulated myosin light-chain kinases (MLCK). However, not all kinases of the MLCK branch are functional MLCKs, and about half lack a CaM binding site in their C-terminal autoinhibitory tail (AI). A unifying feature is their association with the cytoskeleton, mostly via actin and myosin filaments. Titin kinase, similar to its invertebrate analogue twitchin kinase and likely other “MLCKs”, is not Ca2+–calmodulin-activated. Recently, local protein unfolding of the C-terminal AI has emerged as a common mechanism in the activation of CaM kinases. Single-molecule data suggested that opening of the TK active site could also be achieved by mechanical unfolding of the AI. Mechanical modulation of catalytic activity might thus allow cytoskeletal signalling proteins to act as mechanosensors, creating feedback mechanisms between cytoskeletal tension and tension generation or cellular remodelling. Similar to other MLCK-like kinases like DRAK2 and DAPK1, TK is linked to protein turnover regulation via the autophagy/lysosomal system, suggesting the MLCK-like kinases have common functions beyond contraction regulation.
Collapse
Affiliation(s)
- Mathias Gautel
- King's College London BHF Centre of Research Excellence, Cardiovascular Division, London, SE1 1UL, UK.
| |
Collapse
|
16
|
Myosin light chain kinase is central to smooth muscle contraction and required for gastrointestinal motility in mice. Gastroenterology 2008; 135:610-20. [PMID: 18586037 PMCID: PMC2648853 DOI: 10.1053/j.gastro.2008.05.032] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Revised: 04/24/2008] [Accepted: 05/08/2008] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Smooth muscle is essential for maintaining homeostasis for many body functions and provides adaptive responses to stresses imposed by pathologic disorders. Identified cell signaling networks have defined many potential mechanisms for initiating smooth muscle contraction with or without myosin regulatory light chain (RLC) phosphorylation by myosin light chain kinase (MLCK). We generated tamoxifen-inducible and smooth muscle-specific MLCK knockout (KO) mice and provide direct loss-of-function evidence that shows the primary importance of MLCK in phasic smooth muscle contractions. METHODS We used the Cre-loxP system to establish Mlck floxed mice in which exons 23, 24, and 25 were flanked by 2 loxP sites. Smooth muscle-specific MLCK KO mice were generated by crossing Mlck floxed mice with SM-CreER(T2) (ki) mice followed by tamoxifen treatment. The phenotype was assessed by histologic, biochemical, molecular, cell biological, and physiologic analyses. RESULTS Targeted deletion of MLCK in adult mouse smooth muscle resulted in severe gut dysmotility characterized by weak peristalsis, dilation of the digestive tract, and reduction of feces excretion and food intake. There was also abnormal urinary bladder function and lower blood pressure. Isolated muscles showed a loss of RLC phosphorylation and force development induced by K(+)-depolarization. The kinase knockout also markedly reduced RLC phosphorylation and force development with acetylcholine which activates Ca(2+)-sensitizing signaling pathways. CONCLUSIONS MLCK and its phosphorylation of RLC are required physiologically for smooth muscle contraction and are essential for normal gastrointestinal motility.
Collapse
|
17
|
Zhang WC, Peng YJ, He WQ, Lv N, Chen C, Zhi G, Chen HQ, Zhu MS. Identification and functional characterization of an aggregation domain in long myosin light chain kinase. FEBS J 2008; 275:2489-500. [PMID: 18400030 DOI: 10.1111/j.1742-4658.2008.06393.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The functions of long smooth muscle myosin light chain kinase (L-MLCK), a molecule with multiple domains, are poorly understood. To examine the existence of further potentially functional domains in this molecule, we analyzed its amino acid sequence with a tango program and found a putative aggregation domain located at the 4Ig domain of the N-terminal extension. To verify its aggregation capability in vitro, expressible truncated L-MLCK variants driven by a cytomegalovirus promoter were transfected into cells. As anticipated, only the overexpression of the 4Ig fragment led to particle formation in Colon26 cells. These particles contained 4Ig polymers and actin. Analysis with detergents demonstrated that the particles shared features in common with aggregates. Thus, we conclude that the 4Ig domain has a potent aggregation ability. To further examine this aggregation domain in vivo, eight transgenic mouse lines expressing the 4Ig domain (4Ig lines) were generated. The results showed that the transgenic mice had typical aggregation in the thigh and diaphragm muscles. Histological examination showed that 7.70 +/- 1.86% of extensor digitorum longus myofibrils displayed aggregates with a 36.44% reduction in myofibril diameter, whereas 65.13 +/- 3.42% of diaphragm myofibrils displayed aggregates and the myofibril diameter was reduced by 43.08%. Electron microscopy examination suggested that the aggregates were deposited at the mitochondria, resulting in structural impairment. As a consequence, the oxygen consumption of mitochondria in the affected muscles was also reduced. Macrophenotypic analysis showed the presence of muscular degeneration characterized by a reduction in force development, faster fatigue, decreased myofibril diameters, and structural alterations. In summary, our study revealed the existence of a novel aggregation domain in L-MLCK and provided a direct link between L-MLCK and aggregation. The possible significance and mechanism underlying the aggregation-based pathological processes mediated by L-MLCK are also discussed.
Collapse
|
18
|
Funabara D, Hamamoto C, Yamamoto K, Inoue A, Ueda M, Osawa R, Kanoh S, Hartshorne DJ, Suzuki S, Watabe S. Unphosphorylated twitchin forms a complex with actin and myosin that may contribute to tension maintenance in catch. ACTA ACUST UNITED AC 2008; 210:4399-410. [PMID: 18055628 DOI: 10.1242/jeb.008722] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Molluscan smooth muscle can maintain tension over extended periods with little energy expenditure, a process termed catch. Catch is thought to be regulated by phosphorylation of a thick filament protein, twitchin, and involves two phosphorylation sites, D1 and D2, close to the N and C termini, respectively. This study was initiated to investigate the role of the D2 site and its phosphorylation in the catch mechanism. A peptide was constructed containing the D2 site and flanking immunoglobulin (Ig) motifs. It was shown that the dephosphorylated peptide, but not the phosphorylated form, bound to both actin and myosin. The binding site on actin was within the sequence L10 to P29. This region also binds to loop 2 of the myosin head. The dephosphorylated peptide linked myosin and F-actin and formed a trimeric complex. Electron microscopy revealed that twitchin is distributed on the surface of the thick filament with an axial periodicity of 36.25 nm and it is suggested that the D2 site aligns with the myosin heads. It is proposed that the complex formed with the dephosphorylated D2 site of twitchin, F-actin and myosin represents a component of the mechanical linkage in catch.
Collapse
Affiliation(s)
- Daisuke Funabara
- Graduate School of Bioresources, Mie University, Tsu, Mie 514-8507, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Takizawa N, Ikebe R, Ikebe M, Luna EJ. Supervillin slows cell spreading by facilitating myosin II activation at the cell periphery. J Cell Sci 2007; 120:3792-803. [DOI: 10.1242/jcs.008219] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
During cell migration, myosin II modulates adhesion, cell protrusion and actin organization at the leading edge. We show that an F-actin- and membrane-associated scaffolding protein, called supervillin (SV, p205), binds directly to the subfragment 2 domains of nonmuscle myosin IIA and myosin IIB and to the N-terminus of the long form of myosin light chain kinase (L-MLCK). SV inhibits cell spreading via an MLCK- and myosin II-dependent mechanism. Overexpression of SV reduces the rate of cell spreading, and RNAi-mediated knockdown of endogenous SV increases it. Endogenous and EGFP-tagged SV colocalize with, and enhance the formation of, cortical bundles of F-actin and activated myosin II during early cell spreading. The effects of SV are reversed by inhibition of myosin heavy chain (MHC) ATPase (blebbistatin), MLCK (ML-7) or MEK (U0126), but not by inhibiting Rho-kinase with Y-27632. Flag-tagged L-MLCK co-localizes in cortical bundles with EGFP-SV, and kinase-dead L-MLCK disorganizes these bundles. The L-MLCK- and myosin-binding site in SV, SV1-171, rearranges and co-localizes with mono- and di-phosphorylated myosin light chain and with L-MLCK, but not with the short form of MLCK (S-MLCK) or with myosin phosphatase. Thus, the membrane protein SV apparently contributes to myosin II assembly during cell spreading by modulating myosin II regulation by L-MLCK.
Collapse
Affiliation(s)
- Norio Takizawa
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
- Cell Dynamics Program, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Reiko Ikebe
- Department of Physiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
- Cell Dynamics Program, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Mitsuo Ikebe
- Department of Physiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
- Cell Dynamics Program, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Elizabeth J. Luna
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
- Cell Dynamics Program, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
20
|
Yuan J, Shi GX, Shao Y, Dai G, Wei JN, Chang DC, Li CJ. Calmodulin bound to stress fibers but not microtubules involves regulation of cell morphology and motility. Int J Biochem Cell Biol 2007; 40:284-93. [PMID: 17884685 DOI: 10.1016/j.biocel.2007.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Revised: 07/28/2007] [Accepted: 08/05/2007] [Indexed: 01/11/2023]
Abstract
Calmodulin (CaM) is a major cytoplasmic calcium receptor that performs multiple functions including cell motility. To investigate the mechanism of the regulation of CaM on cell morphology and motility, first we checked the distribution of CaM in the living cells using GFP-CaM as an indicator. We found that GFP-CaM showed a fiber-like distribution pattern in the cytosol of living Potorous tridactylis kidney (PtK2) cells but not in living HeLa cells. The endogenous CaM in heavily permeabilized HeLa was also found to display a fiber-like distribution pattern. Further examination showed that the distribution pattern of GFP-CaM was same as that of stress fibers, but not microtubules. Co-immunoprecipitation also showed that CaM can interact with actin directly or indirectly. The microinjection of trp peptide, a specific inhibitor of CaM, attenuated the polymerization of stress fibers and induced the alteration of cell morphology. A wound-healing assay and a single cell tracking experiment showed that CaM in PtK2 cells could increase cell motility. The data we have got from living cells suggested that CaM affect cell morphology and motility through binding to stress fibers and regulate f-actin polymerization.
Collapse
Affiliation(s)
- Jun Yuan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Nanjing Normal University, #1 Wen Yuan Road, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|