1
|
Kyriazi D, Voth L, Bader A, Ewert W, Gerlach J, Elfrink K, Franz P, Tsap MI, Schirmer B, Damiano-Guercio J, Hartmann FK, Plenge M, Salari A, Schöttelndreier D, Strienke K, Bresch N, Salinas C, Gutzeit HO, Schaumann N, Hussein K, Bähre H, Brüsch I, Claus P, Neumann D, Taft MH, Shcherbata HR, Ngezahayo A, Bähler M, Amiri M, Knölker HJ, Preller M, Tsiavaliaris G. An allosteric inhibitor of RhoGAP class-IX myosins suppresses the metastatic features of cancer cells. Nat Commun 2024; 15:9947. [PMID: 39550360 PMCID: PMC11569205 DOI: 10.1038/s41467-024-54181-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 11/01/2024] [Indexed: 11/18/2024] Open
Abstract
Aberrant Ras homologous (Rho) GTPase signalling is a major driver of cancer metastasis, and GTPase-activating proteins (GAPs), the negative regulators of RhoGTPases, are considered promising targets for suppressing metastasis, yet drug discovery efforts have remained elusive. Here, we report the identification and characterization of adhibin, a synthetic allosteric inhibitor of RhoGAP class-IX myosins that abrogates ATPase and motor function, suppressing RhoGTPase-mediated modes of cancer cell metastasis. In human and murine adenocarcinoma and melanoma cell models, including three-dimensional spheroid cultures, we reveal anti-migratory and anti-adhesive properties of adhibin that originate from local disturbances in RhoA/ROCK-regulated signalling, affecting actin-dynamics and actomyosin-based cell-contractility. Adhibin blocks membrane protrusion formation, disturbs remodelling of cell-matrix adhesions, affects contractile ring formation, and disrupts epithelial junction stability; processes severely impairing single/collective cell migration and cytokinesis. Combined with the non-toxic, non-pathological signatures of adhibin validated in organoids, mouse and Drosophila models, this mechanism of action provides the basis for developing anti-metastatic cancer therapies.
Collapse
Affiliation(s)
- Despoina Kyriazi
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Lea Voth
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Almke Bader
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Wiebke Ewert
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
- Institute for Functional Gene Analytics (IFGA), Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | | | - Kerstin Elfrink
- Institute of Integrative Cell Biology and Physiology, University of Münster, Münster, Germany
| | - Peter Franz
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Mariana I Tsap
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Bastian Schirmer
- Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | | | - Falk K Hartmann
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Masina Plenge
- Department of Cell Physiology and Biophysics, Institute of Cell Biology and Biophysics, Leibniz Universität Hannover, Hannover, Germany
| | - Azam Salari
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | | | - Katharina Strienke
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Nadine Bresch
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Claudio Salinas
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | | | - Nora Schaumann
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - Kais Hussein
- Institute of Pathology, KRH Klinikum Nordstadt, Hannover, Germany
| | - Heike Bähre
- Research Core Unit Mass Spectrometry-Metabolomics, Hannover Medical School, Hanover, Germany
| | - Inga Brüsch
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Peter Claus
- SMATHERIA gGmbH-Non-Profit Biomedical Research Institute, Hannover, Germany
| | - Detlef Neumann
- Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | - Manuel H Taft
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | | | - Anaclet Ngezahayo
- Department of Cell Physiology and Biophysics, Institute of Cell Biology and Biophysics, Leibniz Universität Hannover, Hannover, Germany
| | - Martin Bähler
- Institute of Integrative Cell Biology and Physiology, University of Münster, Münster, Germany
| | - Mahdi Amiri
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | | | - Matthias Preller
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
- Institute for Functional Gene Analytics (IFGA), Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | | |
Collapse
|
2
|
El-Mansi S, Mitchell TP, Mobayen G, McKinnon TAJ, Miklavc P, Frick M, Nightingale TD. Myosin-1C augments endothelial secretion of von Willebrand factor by linking contractile actomyosin machinery to the plasma membrane. Blood Adv 2024; 8:4714-4726. [PMID: 38669344 PMCID: PMC11413703 DOI: 10.1182/bloodadvances.2024012590] [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: 01/08/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
ABSTRACT Blood endothelial cells control the hemostatic and inflammatory response by secreting von Willebrand factor (VWF) and P-selectin from storage organelles called Weibel-Palade bodies (WPBs). Actin-associated motor proteins regulate this secretory pathway at multiple points. Before fusion, myosin Va forms a complex that anchors WPBs to peripheral actin structures, allowing for the maturation of content. After fusion, an actomyosin ring/coat is recruited and compresses the WPB to forcibly expel the largest VWF multimers. Here, we provide, to our knowledge, the first evidence for the involvement of class I myosins during regulated VWF secretion. We show that the unconventional myosin-1C (Myo1c) is recruited after fusion via its pleckstrin homology domain in an actin-independent process. This provides a link between the actin ring and phosphatidylinositol 4,5-bisphosphate (PIP2) at the membrane of the fused organelle and is necessary to ensure maximal VWF secretion. This is an active process requiring Myo1c ATPase activity because inhibition of class I myosins using the inhibitor pentachloropseudilin or expression of an ATPase-deficient Myo1c rigor mutant perturbs the expulsion of VWF and alters the kinetics of the exocytic actin ring. These data offer a novel insight into the control of an essential physiological process and provide a new way in which it can be regulated.
Collapse
Affiliation(s)
- Sammy El-Mansi
- Centre for Microvascular Research, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Tom P. Mitchell
- Centre for Microvascular Research, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Golzar Mobayen
- Department of Immunology and Inflammation, Centre for Haematology, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Thomas A. J. McKinnon
- Department of Immunology and Inflammation, Centre for Haematology, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Pika Miklavc
- School of Science, Engineering & Environment, University of Salford, Manchester, United Kingdom
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Thomas D. Nightingale
- Centre for Microvascular Research, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
3
|
Castro-Falcón G, Straetener J, Bornikoel J, Reimer D, Purdy TN, Berscheid A, Schempp FM, Liu DY, Linington RG, Brötz-Oesterhelt H, Hughes CC. Antibacterial Marinopyrroles and Pseudilins Act as Protonophores. ACS Chem Biol 2024; 19:743-752. [PMID: 38377384 PMCID: PMC10949930 DOI: 10.1021/acschembio.3c00773] [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: 12/15/2023] [Accepted: 12/27/2023] [Indexed: 02/22/2024]
Abstract
Elucidating the mechanism of action (MoA) of antibacterial natural products is crucial to evaluating their potential as novel antibiotics. Marinopyrroles, pentachloropseudilin, and pentabromopseudilin are densely halogenated, hybrid pyrrole-phenol natural products with potent activity against Gram-positive bacterial pathogens like Staphylococcus aureus. However, the exact way they exert this antibacterial activity has not been established. In this study, we explore their structure-activity relationship, determine their spatial location in bacterial cells, and investigate their MoA. We show that the natural products share a common MoA based on membrane depolarization and dissipation of the proton motive force (PMF) that is essential for cell viability. The compounds show potent protonophore activity but do not appear to destroy the integrity of the cytoplasmic membrane via the formation of larger pores or interfere with the stability of the peptidoglycan sacculus. Thus, our current model for the antibacterial MoA of marinopyrrole, pentachloropseudilin, and pentabromopseudilin stipulates that the acidic compounds insert into the membrane and transport protons inside the cell. This MoA may explain many of the deleterious biological effects in mammalian cells, plants, phytoplankton, viruses, and protozoans that have been reported for these compounds.
Collapse
Affiliation(s)
- Gabriel Castro-Falcón
- Center
for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California, San Diego, California 92093, United States
| | - Jan Straetener
- Department
of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology
and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
| | - Jan Bornikoel
- Department
of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology
and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
| | - Daniela Reimer
- Center
for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California, San Diego, California 92093, United States
| | - Trevor N. Purdy
- Center
for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California, San Diego, California 92093, United States
| | - Anne Berscheid
- Department
of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology
and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
| | - Florence M. Schempp
- Center
for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California, San Diego, California 92093, United States
| | - Dennis Y. Liu
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Roger G. Linington
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Heike Brötz-Oesterhelt
- Department
of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology
and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
- Cluster
of Excellence EXC 2124: Controlling Microbes to Fight Infection, University of Tübingen, Tübingen 72076, Germany
- German
Center for Infection Research, Partner Site Tübingen, Tübingen 72076, Germany
| | - Chambers C. Hughes
- Center
for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California, San Diego, California 92093, United States
- Department
of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology
and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
- Cluster
of Excellence EXC 2124: Controlling Microbes to Fight Infection, University of Tübingen, Tübingen 72076, Germany
- German
Center for Infection Research, Partner Site Tübingen, Tübingen 72076, Germany
| |
Collapse
|
4
|
Liu C, Ruppel KM, Spudich JA. Motility Assay to Probe the Calcium Sensitivity of Myosin and Regulated Thin Filaments. Methods Mol Biol 2024; 2735:169-189. [PMID: 38038849 PMCID: PMC10773985 DOI: 10.1007/978-1-0716-3527-8_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Calcium-dependent activation of the thin filament mediated by the troponin-tropomyosin complex is key in the regulation of actin-myosin based muscle contraction. Perturbations to this system, either physiological (e.g., phosphorylation of myosin light chains) or pathological (e.g., mutations that cause familial cardiomyopathies), can alter calcium sensitivity and thus have important implications in human health and disease. The in vitro motility assay provides a quantitative and precise method to study the calcium sensitivity of the reconstituted myosin-thin filament motile system. Here we present a simple and robust protocol to perform calcium-dependent motility of β-cardiac myosin and regulated thin filaments. The experiment is done on a multichannel microfluidic slide requiring minimal amounts of proteins. A complete velocity vs. calcium concentration curve is produced from one experiment in under 1 h.
Collapse
Affiliation(s)
- Chao Liu
- Department of Biochemistry, Beckman Center B405, Stanford University School of Medicine, Stanford, CA, USA
- Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Kathleen M Ruppel
- Department of Biochemistry, Beckman Center B405, Stanford University School of Medicine, Stanford, CA, USA.
- Cardiovascular Institute, Stanford University, Stanford, CA, USA.
- Department of Pediatrics (Cardiology), Stanford University, Stanford, CA, USA.
| | - James A Spudich
- Department of Biochemistry, Beckman Center B405, Stanford University School of Medicine, Stanford, CA, USA.
- Cardiovascular Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
5
|
Burguera S, Frontera A, Bauzá A. Biological noncovalent N/O⋯V interactions: insights from theory and protein data bank analyses. Phys Chem Chem Phys 2023; 25:30040-30048. [PMID: 37905702 DOI: 10.1039/d3cp04571d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Computations at the PBE0-D3/def2-TZVP level of theory in conjunction with a Protein Data Bank (PDB) survey have provided first time evidence of favorable noncovalent interactions between ADP metavanadate (VO4) and ADP orthovanadate (VO5) and electron rich atoms. These involve a σ-hole present in the V atom and the lone pairs belonging to (i) protein residues (e.g., serine (SER), glutamate (GLU) or histidine (HIS)), (ii) backbone carbonyl groups and (iii) water molecules. A computational study has been carried out to rationalize the physical nature and directionality of the interaction in addition to its plausible biological role. The results reported herein are expected to have an impact in the fields of medicinal chemistry, bioinorganic chemistry and chemical biology.
Collapse
Affiliation(s)
- Sergi Burguera
- Department of Chemistry, Universitat de les Illes Balears, Ctra. de Valldemossa km. 7.5, 07122, Palma de Mallorca, Islas Baleares, Spain.
| | - Antonio Frontera
- Department of Chemistry, Universitat de les Illes Balears, Ctra. de Valldemossa km. 7.5, 07122, Palma de Mallorca, Islas Baleares, Spain.
| | - Antonio Bauzá
- Department of Chemistry, Universitat de les Illes Balears, Ctra. de Valldemossa km. 7.5, 07122, Palma de Mallorca, Islas Baleares, Spain.
| |
Collapse
|
6
|
Zheng W. Predicting allosteric sites using fast conformational sampling as guided by coarse-grained normal modes. J Chem Phys 2023; 158:124127. [PMID: 37003737 PMCID: PMC10066797 DOI: 10.1063/5.0141630] [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: 01/07/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023] Open
Abstract
To computationally identify cryptic binding sites for allosteric modulators, we have developed a fast and simple conformational sampling scheme guided by coarse-grained normal modes solved from the elastic network models followed by atomistic backbone and sidechain reconstruction. Despite the complexity of conformational changes associated with ligand binding, we previously showed that simply sampling along each of the lowest 30 modes can adequately restructure cryptic sites so they are detectable by pocket finding programs like Concavity. Here, we applied this method to study four classical examples of allosteric regulation (GluR2 receptor, GroEL chaperonin, GPCR, and myosin). Our method along with alternative methods has been utilized to locate known allosteric sites and predict new promising allosteric sites. Compared with other sampling methods based on extensive molecular dynamics simulation, our method is both faster (1-2 h for an average-size protein of ∼400 residues) and more flexible (it can be easily integrated with any structure-based pocket finding methods), so it is suitable for high-throughput screening of large datasets of protein structures at the genome scale.
Collapse
Affiliation(s)
- Wenjun Zheng
- Department of Physics, University at Buffalo, 239 Fronczak Hall, Buffalo, New York 14260, USA
| |
Collapse
|
7
|
Spirandelli da Costa M, Borges BC, Marques IT, de Oliveira RC, Teixeira TL, de Gouveia Santos J, Silva CVD. Pentachloropseudilin treatment impairs host cell invasion by Trypanosoma cruzi. Chembiochem 2022; 23:e202200349. [PMID: 35839379 DOI: 10.1002/cbic.202200349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/14/2022] [Indexed: 11/08/2022]
Abstract
Pentachloropseudilin (PClP) is a reversible and allosteric inhibitor of type 1 myosin. Here, we addressed the impact of PClP treatment of Trypanosoma cruzi and mammalian host cell on the parasite migration, cell adhesion and invasion. We observed that PClP was not toxic to either T. cruzi or host cell. Moreover, treatment of T. cruzi with PClP inhbited parasite motility, host cell adhesion and invasion. Treatment of host cell with PClP also impaired parasite invasion probably by decreasing lysosome migration to the entry site of the parasite. Therefore, PClP treatment impaired fundamental processes necessary for a successful T. cruzi infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Claudio Vieira da Silva
- Universidade Federal de Uberlândia, Imunologia, Rua Piauí, Bloco 2B sala 200, 38400096, Uberlândia, BRAZIL
| |
Collapse
|
8
|
Radhakrishnan R, Dronamraju VR, Leung M, Gruesen A, Solanki AK, Walterhouse S, Roehrich H, Song G, da Costa Monsanto R, Cureoglu S, Martin R, Kondkar AA, van Kuijk FJ, Montezuma SR, Knöelker HJ, Hufnagel RB, Lobo GP. The role of motor proteins in photoreceptor protein transport and visual function. Ophthalmic Genet 2022; 43:285-300. [PMID: 35470760 DOI: 10.1080/13816810.2022.2062391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Rods and cones are photoreceptor neurons in the retina that are required for visual sensation in vertebrates, wherein the perception of vision is initiated when these neurons respond to photons in the light stimuli. The photoreceptor cell is structurally studied as outer segments (OS) and inner segments (IS) where proper protein sorting, localization, and compartmentalization are critical for phototransduction, visual function, and survival. In human retinal diseases, improper protein transport to the OS or mislocalization of proteins to the IS and other cellular compartments could lead to impaired visual responses and photoreceptor cell degeneration that ultimately cause loss of visual function. RESULTS Therefore, studying and identifying mechanisms involved in facilitating and maintaining proper protein transport in photoreceptor cells would help our understanding of pathologies involving retinal cell degeneration in inherited retinal dystrophies, age-related macular degeneration, and Usher Syndrome. CONCLUSIONS Our mini-review will discuss mechanisms of protein transport within photoreceptors and introduce a novel role for an unconventional motor protein, MYO1C, in actin-based motor transport of the visual chromophore Rhodopsin to the OS, in support of phototransduction and visual function.
Collapse
Affiliation(s)
- Rakesh Radhakrishnan
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Venkateshwara R Dronamraju
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Matthias Leung
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Andrew Gruesen
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ashish K Solanki
- Department of Medicine, Drug Discovery Building, Medical University of South Carolina, South Carolina, USA
| | - Stephen Walterhouse
- Department of Medicine, Drug Discovery Building, Medical University of South Carolina, South Carolina, USA
| | - Heidi Roehrich
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Grace Song
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rafael da Costa Monsanto
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sebahattin Cureoglu
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - René Martin
- Faculty of Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Altaf A Kondkar
- Department of Ophthalmology.,Glaucoma Research Chair in Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Frederik J van Kuijk
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sandra R Montezuma
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Robert B Hufnagel
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Glenn P Lobo
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Medicine, Drug Discovery Building, Medical University of South Carolina, South Carolina, USA.,Department of Ophthalmology, Medical University of South Carolina, South Carolina, USA
| |
Collapse
|
9
|
Wang Y, Zhao X, Gao M, Wan X, Guo Y, Qu Y, Chen Y, Li T, Liu H, Jiang M, Wang F, Sun X. Myosin 1f-mediated activation of microglia contributes to the photoreceptor degeneration in a mouse model of retinal detachment. Cell Death Dis 2021; 12:926. [PMID: 34628463 PMCID: PMC8502177 DOI: 10.1038/s41419-021-03983-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 11/14/2022]
Abstract
Photoreceptor death and neurodegeneration is the leading cause of irreversible vision loss. The inflammatory response of microglia plays an important role in the process of neurodegeneration. In this study, we chose retinal detachment as the model of photoreceptor degeneration. We found Myosin 1f was upregulated after retinal detachment, and it was specifically expressed in microglia. Deficiency of myosin 1f protected against photoreceptor apoptosis by inhibiting microglia activation. The elimination of microglia can abolish the protective effect of myosin 1f deficiency. After stimulation by LPS, microglia with myosin 1f deficiency showed downregulation of the MAPK and AKT pathways. Our results demonstrated that myosin 1f plays a crucial role in microglia-induced neuroinflammation after retinal injury and photoreceptor degeneration by regulating two classic inflammatory pathways and thereby decreasing the expression of inflammatory cytokines. Knockout of myosin 1f reduces the intensity of the immune response and prevents cell death of photoreceptor, suggesting that myosin 1f can be inhibited to prevent a decline in visual acuity after retinal detachment.
Collapse
Affiliation(s)
- Yimin Wang
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Xiaohuan Zhao
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Min Gao
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoling Wan
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Yinong Guo
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Yingying Qu
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Immunology, Translational Medicine Center, Shanghai General Hospital, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Tong Li
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
| | - Haiyun Liu
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
| | - Mei Jiang
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Disease, Shanghai, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China
| | - Feng Wang
- Shanghai Institute of Immunology, Translational Medicine Center, Shanghai General Hospital, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiaodong Sun
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- National Clinical Research Center for Eye Disease, Shanghai, China.
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, China.
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.
| |
Collapse
|
10
|
Arif E, Wang C, Swiderska-Syn MK, Solanki AK, Rahman B, Manka PP, Coombes JD, Canbay A, Papa S, Nihalani D, Aspichueta P, Lipschutz JH, Syn WK. Targeting myosin 1c inhibits murine hepatic fibrogenesis. Am J Physiol Gastrointest Liver Physiol 2021; 320:G1044-G1053. [PMID: 33908271 PMCID: PMC8285590 DOI: 10.1152/ajpgi.00105.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Myosin 1c (Myo1c) is an unconventional myosin that modulates signaling pathways involved in tissue injury and repair. In this study, we observed that Myo1c expression is significantly upregulated in human chronic liver disease such as nonalcoholic steatohepatitis (NASH) and in animal models of liver fibrosis. High throughput data from the GEO-database identified similar Myo1c upregulation in mice and human liver fibrosis. Notably, transforming growth factor-β1 (TGF-β1) stimulation to hepatic stellate cells (HSCs), the liver pericyte and key cell type responsible for the deposition of extracellular matrix, upregulates Myo1c expression, whereas genetic depletion or pharmacological inhibition of Myo1c blunted TGF-β-induced fibrogenic responses, resulting in repression of α-smooth muscle actin (α-SMA) and collagen type I α 1 chain (Col1α1) mRNA. Myo1c deletion also decreased fibrogenic processes such as cell proliferation, wound healing response, and contractility when compared with vehicle-treated HSCs. Importantly, phosphorylation of mothers against decapentaplegic homolog 2 (SMAD2) and mothers against decapentaplegic homolog 3 (SMAD3) were significantly blunted upon Myo1c inhibition in GRX cells as well as Myo1c knockout (Myo1c-KO) mouse embryonic fibroblasts (MEFs) upon TGF-β stimulation. Using the genetic Myo1c-KO mice, we confirmed that Myo1c is critical for fibrogenesis, as Myo1c-KO mice were resistant to carbon tetrachloride (CCl4)-induced liver fibrosis. Histological and immunostaining analysis of liver sections showed that deposition of collagen fibers and α-SMA expression were significantly reduced in Myo1c-KO mice upon liver injury. Collectively, these results demonstrate that Myo1c mediates hepatic fibrogenesis by modulating TGF-β signaling and suggest that inhibiting this process may have clinical application in treating liver fibrosis.NEW & NOTEWORTHY The incidences of liver fibrosis are growing at a rapid pace and have become one of the leading causes of end-stage liver disease. Although TGF-β1 is known to play a prominent role in transforming cells to produce excessive extracellular matrix that lead to hepatic fibrosis, the therapies targeting TGF-β1 have achieved very limited clinical impact. This study highlights motor protein myosin-1c-mediated mechanisms that serve as novel regulators of TGF-β1 signaling and fibrosis.
Collapse
Affiliation(s)
- Ehtesham Arif
- 1Department of Medicine, Nephrology Division, Medical University of South Carolinagrid.259828.c, Charleston, South Carolina,2Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, South Carolina
| | - Cindy Wang
- 2Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, South Carolina
| | - Marzena K. Swiderska-Syn
- 3Department of Pediatrics, Darby Children’s Research Institute,
Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Ashish K. Solanki
- 1Department of Medicine, Nephrology Division, Medical University of South Carolinagrid.259828.c, Charleston, South Carolina
| | - Bushra Rahman
- 1Department of Medicine, Nephrology Division, Medical University of South Carolinagrid.259828.c, Charleston, South Carolina
| | - Paul P. Manka
- 2Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, South Carolina,4Department of Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, Bochum, Germany
| | - Jason D. Coombes
- 5Institute of Hepatology, Foundation for Liver Research, London, United Kingdom,6School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Ali Canbay
- 4Department of Medicine, University Hospital Knappschaftskrankenhaus, Ruhr-University Bochum, Bochum, Germany
| | - Salvatore Papa
- 7Leeds Institute of Medical Research at St. James’s, Faculty of
Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Deepak Nihalani
- 1Department of Medicine, Nephrology Division, Medical University of South Carolinagrid.259828.c, Charleston, South Carolina,8Division of Kidney, Urologic and Hematologic Diseases, National Institutes of Health, Bethesda, Maryland
| | - Patricia Aspichueta
- 9Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Leioa, Spain
| | - Joshua H. Lipschutz
- 1Department of Medicine, Nephrology Division, Medical University of South Carolinagrid.259828.c, Charleston, South Carolina,10Section of Nephrology, Ralph H Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Wing-Kin Syn
- 2Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, South Carolina,9Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Leioa, Spain,11Section of Gastroenterology, Ralph H Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
11
|
Åslund A, Bokhari MH, Wetterdal E, Martin R, Knölker HJ, Bengtsson T. Myosin 1c: A novel regulator of glucose uptake in brown adipocytes. Mol Metab 2021; 53:101247. [PMID: 33965643 PMCID: PMC8182130 DOI: 10.1016/j.molmet.2021.101247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/26/2021] [Accepted: 05/03/2021] [Indexed: 12/02/2022] Open
Abstract
Objective The potential of brown adipose tissue (BAT) to influence energy homeostasis in animals and humans is encouraging as this tissue can increase fatty acid and glucose utilization to produce heat through uncoupling protein 1 (UCP1), but the actual mechanism of how the cell regulates glucose uptake is not fully understood. Myosin 1c (Myo1c) is an unconventional motor protein involved in several cellular processes, including insulin-mediated glucose uptake via GLUT4 vesicle fusion in white adipocytes, but its role in glucose uptake in BAT has not previously been investigated. Methods Using the specific inhibitor pentachloropseudilin (PClP), a neutralizing antibody assay, and siRNA, we examined the role of Myo1c in mechanisms leading to glucose uptake both in vitro in isolated mouse primary adipocytes and in vivo in mice. Results Our results show that inhibition of Myo1c removes insulin-stimulated glucose uptake in white adipocytes, while inducing glucose uptake in brown adipocytes, independent of GLUT4, by increasing the expression, translation, and translocation of GLUT1 to the plasma membrane. Inhibition of Myo1c leads to the activation of PKA and downstream substrates p38 and ATF-2, which are known to be involved in the expression of β-adrenergic genes. Conclusions Myo1c is a PKA repressor and regulates glucose uptake into BAT. Myo1c is a BAT-specific regulator of glucose uptake. Myo1c inhibition leads to increased expression, translation, and translocation of GLUT1. Myo1c inhibition results in increased activation of PKA and its downstream targets.
Collapse
Affiliation(s)
- Alice Åslund
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, SE-106 91, Stockholm, Sweden
| | - Muhammad Hamza Bokhari
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, SE-106 91, Stockholm, Sweden
| | - Erika Wetterdal
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, SE-106 91, Stockholm, Sweden
| | - René Martin
- Faculty of Chemistry, Technical University of Dresden, Bergstrasse 66, 01069, Dresden, Germany
| | - Hans-Joachim Knölker
- Faculty of Chemistry, Technical University of Dresden, Bergstrasse 66, 01069, Dresden, Germany
| | - Tore Bengtsson
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, SE-106 91, Stockholm, Sweden.
| |
Collapse
|
12
|
Tokuo H, Komaba S, Coluccio LM. In pancreatic β-cells myosin 1b regulates glucose-stimulated insulin secretion by modulating an early step in insulin granule trafficking from the Golgi. Mol Biol Cell 2021; 32:1210-1220. [PMID: 33826361 PMCID: PMC8351557 DOI: 10.1091/mbc.e21-03-0094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Pancreatic β-cells secrete insulin, which controls blood glucose levels, and defects in insulin secretion are responsible for diabetes mellitus. The actin cytoskeleton and some myosins support insulin granule trafficking and release, although a role for the class I myosin Myo1b, an actin- and membrane-associated load-sensitive motor, in insulin biology is unknown. We found by immunohistochemistry that Myo1b is expressed in islet cells of the rat pancreas. In cultured rat insulinoma 832/13 cells, Myo1b localized near actin patches, the trans-Golgi network (TGN) marker TGN38, and insulin granules in the perinuclear region. Myo1b depletion by small interfering RNA in 832/13 cells reduced intracellular proinsulin and insulin content and glucose-stimulated insulin secretion (GSIS) and led to the accumulation of (pro)insulin secretory granules (SGs) at the TGN. Using an in situ fluorescent pulse-chase strategy to track nascent proinsulin, Myo1b depletion in insulinoma cells reduced the number of (pro)insulin-containing SGs budding from the TGN. The studies indicate for the first time that in pancreatic β-cells Myo1b controls GSIS at least in part by mediating an early stage in insulin granule trafficking from the TGN.
Collapse
Affiliation(s)
- Hiroshi Tokuo
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA 02118-2518
| | - Shigeru Komaba
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA 02118-2518
| | - Lynne M Coluccio
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA 02118-2518
| |
Collapse
|
13
|
Ruggiero C, Lalli E. Targeting the cytoskeleton against metastatic dissemination. Cancer Metastasis Rev 2021; 40:89-140. [PMID: 33471283 DOI: 10.1007/s10555-020-09936-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 10/08/2020] [Indexed: 02/08/2023]
Abstract
Cancer is a pathology characterized by a loss or a perturbation of a number of typical features of normal cell behaviour. Indeed, the acquisition of an inappropriate migratory and invasive phenotype has been reported to be one of the hallmarks of cancer. The cytoskeleton is a complex dynamic network of highly ordered interlinking filaments playing a key role in the control of fundamental cellular processes, like cell shape maintenance, motility, division and intracellular transport. Moreover, deregulation of this complex machinery contributes to cancer progression and malignancy, enabling cells to acquire an invasive and metastatic phenotype. Metastasis accounts for 90% of death from patients affected by solid tumours, while an efficient prevention and suppression of metastatic disease still remains elusive. This results in the lack of effective therapeutic options currently available for patients with advanced disease. In this context, the cytoskeleton with its regulatory and structural proteins emerges as a novel and highly effective target to be exploited for a substantial therapeutic effort toward the development of specific anti-metastatic drugs. Here we provide an overview of the role of cytoskeleton components and interacting proteins in cancer metastasis with a special focus on small molecule compounds interfering with the actin cytoskeleton organization and function. The emerging involvement of microtubules and intermediate filaments in cancer metastasis is also reviewed.
Collapse
Affiliation(s)
- Carmen Ruggiero
- Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, CNRS, 660 route des Lucioles-Sophia Antipolis, 06560, Valbonne, France.
- NEOGENEX-CANCER CNRS International Associated Laboratory, 660 route des Lucioles, Sophia Antipolis, 06560, Valbonne, France.
| | - Enzo Lalli
- NEOGENEX-CANCER CNRS International Associated Laboratory, 660 route des Lucioles, Sophia Antipolis, 06560, Valbonne, France
- Inserm, Institut de Pharmacologie Moléculaire et Cellulaire, 660 route des Lucioles - Sophia Antipolis, 06560, Valbonne, France
| |
Collapse
|
14
|
Naydenov NG, Lechuga S, Huang EH, Ivanov AI. Myosin Motors: Novel Regulators and Therapeutic Targets in Colorectal Cancer. Cancers (Basel) 2021; 13:741. [PMID: 33670106 PMCID: PMC7916823 DOI: 10.3390/cancers13040741] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) remains the third most common cause of cancer and the second most common cause of cancer deaths worldwide. Clinicians are largely faced with advanced and metastatic disease for which few interventions are available. One poorly understood aspect of CRC involves altered organization of the actin cytoskeleton, especially at the metastatic stage of the disease. Myosin motors are crucial regulators of actin cytoskeletal architecture and remodeling. They act as mechanosensors of the tumor environments and control key cellular processes linked to oncogenesis, including cell division, extracellular matrix adhesion and tissue invasion. Different myosins play either oncogenic or tumor suppressor roles in breast, lung and prostate cancer; however, little is known about their functions in CRC. This review focuses on the functional roles of myosins in colon cancer development. We discuss the most studied class of myosins, class II (conventional) myosins, as well as several classes (I, V, VI, X and XVIII) of unconventional myosins that have been linked to CRC development. Altered expression and mutations of these motors in clinical tumor samples and their roles in CRC growth and metastasis are described. We also evaluate the potential of using small molecular modulators of myosin activity to develop novel anticancer therapies.
Collapse
Affiliation(s)
- Nayden G. Naydenov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (N.G.N.); (S.L.)
| | - Susana Lechuga
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (N.G.N.); (S.L.)
| | - Emina H. Huang
- Departments of Cancer Biology and Colorectal Surgery, Cleveland Clinic Foundation, Cleveland, OH 44195, USA;
| | - Andrei I. Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (N.G.N.); (S.L.)
| |
Collapse
|
15
|
Richter S, Martin R, Gutzeit HO, Knölker HJ. In vitro and in vivo effects of inhibitors on actin and myosin. Bioorg Med Chem 2021; 30:115928. [PMID: 33341499 DOI: 10.1016/j.bmc.2020.115928] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/30/2020] [Accepted: 12/03/2020] [Indexed: 11/18/2022]
Abstract
The interaction of actin and myosin is essential for cell migration. We have identified kaempferol and pentahalogenated pseudilins as efficient inhibitors of migration of MDA-MB-231 breast adenocarcinoma cells. The compounds were studied with respect to possible effects on myosin-2-ATPase activity. The pentahalogenated pseudilins inhibited the enzyme activity in vitro. Flavonoids showed no effect on enzyme activity. The polymerization dynamics of actin was measured to test whether the integrity of F-actin is essential for the migration of MDA-MB-231 cells. Quercetin and kaempferol depolymerized F-actin with similar efficiencies as found for the pentahalogenated pseudilins, whereas epigallocatechin showed the weakest effect. As the inhibitory effect on cell migration may be caused by a toxic effect, we have performed a cytotoxicity test and, furthermore, investigated the influence of the test compounds on cardiac function in eleutheroembryos of medaka (Oryzias latipes). Compared with the pentahalogenated pseudilins, the cytotoxic and cardiotoxic effects of flavonoids on medaka embryos were found to be moderate.
Collapse
Affiliation(s)
- Sabine Richter
- Faculty of Biology, TU Dresden, Zellescher Weg 20b, 01217 Dresden, Germany
| | - René Martin
- Faculty of Chemistry, TU Dresden, Bergstrasse 66, 01069 Dresden, Germany
| | - Herwig O Gutzeit
- Faculty of Biology, TU Dresden, Zellescher Weg 20b, 01217 Dresden, Germany.
| | | |
Collapse
|
16
|
Undefeated-Changing the phenamacril scaffold is not enough to beat resistant Fusarium. PLoS One 2020; 15:e0235568. [PMID: 32598376 PMCID: PMC7323951 DOI: 10.1371/journal.pone.0235568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/17/2020] [Indexed: 11/19/2022] Open
Abstract
Filamentous fungi belonging to the genus Fusarium are notorious plant-pathogens that infect, damage and contaminate a wide variety of important crops. Phenamacril is the first member of a novel class of single-site acting cyanoacrylate fungicides which has proven highly effective against important members of the genus Fusarium. However, the recent emergence of field-resistant strains exhibiting qualitative resistance poses a major obstacle for the continued use of phenamacril. In this study, we synthesized novel cyanoacrylate compounds based on the phenamacril-scaffold to test their growth-inhibitory potential against wild-type Fusarium and phenamacril-resistant strains. Our findings show that most chemical modifications to the phenamacril-scaffold are associated with almost complete loss of fungicidal activity and in vitro inhibition of myosin motor domain ATPase activity.
Collapse
|
17
|
Drug Discovery against Acanthamoeba Infections: Present Knowledge and Unmet Needs. Pathogens 2020; 9:pathogens9050405. [PMID: 32456110 PMCID: PMC7281112 DOI: 10.3390/pathogens9050405] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/09/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022] Open
Abstract
Although major strides have been made in developing and testing various anti-acanthamoebic drugs, recurrent infections, inadequate treatment outcomes, health complications, and side effects associated with the use of currently available drugs necessitate the development of more effective and safe therapeutic regimens. For any new anti-acanthamoebic drugs to be more effective, they must have either superior potency and safety or at least comparable potency and an improved safety profile compared to the existing drugs. The development of the so-called 'next-generation' anti-acanthamoebic agents to address this challenge is an active area of research. Here, we review the current status of anti-acanthamoebic drugs and discuss recent progress in identifying novel pharmacological targets and new approaches, such as drug repurposing, development of small interfering RNA (siRNA)-based therapies and testing natural products and their derivatives. Some of the discussed approaches have the potential to change the therapeutic landscape of Acanthamoeba infections.
Collapse
|
18
|
Zhou Y, Zhou XE, Gong Y, Zhu Y, Cao X, Brunzelle JS, Xu HE, Zhou M, Melcher K, Zhang F. Structural basis of Fusarium myosin I inhibition by phenamacril. PLoS Pathog 2020; 16:e1008323. [PMID: 32163521 PMCID: PMC7100991 DOI: 10.1371/journal.ppat.1008323] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 03/27/2020] [Accepted: 01/16/2020] [Indexed: 11/26/2022] Open
Abstract
Fusarium is a genus of filamentous fungi that includes species that cause devastating diseases in major staple crops, such as wheat, maize, rice, and barley, resulting in severe yield losses and mycotoxin contamination of infected grains. Phenamacril is a novel fungicide that is considered environmentally benign due to its exceptional specificity; it inhibits the ATPase activity of the sole class I myosin of only a subset of Fusarium species including the major plant pathogens F. graminearum, F. asiaticum and F. fujikuroi. To understand the underlying mechanisms of inhibition, species specificity, and resistance mutations, we have determined the crystal structure of phenamacril-bound F. graminearum myosin I. Phenamacril binds in the actin-binding cleft in a new allosteric pocket that contains the central residue of the regulatory Switch 2 loop and that is collapsed in the structure of a myosin with closed actin-binding cleft, suggesting that pocket occupancy blocks cleft closure. We have further identified a single, transferable phenamacril-binding residue found exclusively in phenamacril-sensitive myosins to confer phenamacril selectivity. Phenamacril is a recently identified myosin I inhibitor that is a potent and highly species-specific and myosin subtype-selective fungicide. We report the high-resolution structure of the phenamacril-bound myosin I motor domain of the major crop pathogen Fusarium graminearum, providing insight into the molecular mechanism of phenamacril action and resistance. These results are of broad significance for understanding the mode of actions of myosin-based fungicides and for designing novel myosin I inhibitors for crop protection and for treatment of human myosin dysfunction diseases.
Collapse
Affiliation(s)
- Yuxin Zhou
- Key Laboratory of Pesticide, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
- Center of Cancer and Cell Biology, Program for Structural Biology, Van Andel Institute, Grand Rapids, Michigan, United States of America
| | - X. Edward Zhou
- Center of Cancer and Cell Biology, Program for Structural Biology, Van Andel Institute, Grand Rapids, Michigan, United States of America
| | - Yuanping Gong
- Key Laboratory of Pesticide, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Yuanye Zhu
- Key Laboratory of Pesticide, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Xiaoman Cao
- Key Laboratory of Pesticide, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Joseph S. Brunzelle
- Northwestern University Synchrotron Research Center, Life Sciences Collaborative Access Team, Northwestern University, Argonne, Illinois, United States of America
| | - H. Eric Xu
- Center of Cancer and Cell Biology, Program for Structural Biology, Van Andel Institute, Grand Rapids, Michigan, United States of America
- Center for Structure and Function of Drug Targets, The CAS-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Mingguo Zhou
- Key Laboratory of Pesticide, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
- * E-mail: (MZ); (KM); (FZ)
| | - Karsten Melcher
- Center of Cancer and Cell Biology, Program for Structural Biology, Van Andel Institute, Grand Rapids, Michigan, United States of America
- * E-mail: (MZ); (KM); (FZ)
| | - Feng Zhang
- Key Laboratory of Pesticide, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
- * E-mail: (MZ); (KM); (FZ)
| |
Collapse
|
19
|
Ferro V, Chuai M, McGloin D, Weijer CJ. Measurement of junctional tension in epithelial cells at the onset of primitive streak formation in the chick embryo via non-destructive optical manipulation. Development 2020; 147:dev.175109. [PMID: 31964776 PMCID: PMC7033729 DOI: 10.1242/dev.175109] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 01/13/2020] [Indexed: 12/23/2022]
Abstract
Directional cell intercalations of epithelial cells during gastrulation has, in several organisms, been shown to be associated with a planar cell polarity in the organisation of the actin-myosin cytoskeleton and is postulated to reflect directional tension that drives oriented cell intercalations. We have characterised and applied a recently introduced non-destructive optical manipulation technique to measure the tension in individual epithelial cell junctions of cells in various locations and orientations in the epiblast of chick embryos in the early stages of primitive streak formation. Junctional tension of mesendoderm precursors in the epiblast is higher in junctions oriented in the direction of intercalation than in junctions oriented perpendicular to the direction of intercalation and higher than in junctions of other cells in the epiblast. The kinetic data fit best with a simple viscoelastic Maxwell model, and we find that junctional tension, and to a lesser extent viscoelastic relaxation time, are dependent on myosin activity.
Collapse
Affiliation(s)
- Valentina Ferro
- Department of Physics, School of Science and Engineering, University of Dundee, Dundee DD1 4HN, UK
| | - Manli Chuai
- School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - David McGloin
- Department of Physics, School of Science and Engineering, University of Dundee, Dundee DD1 4HN, UK
- School of Electrical and Data Engineering, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | | |
Collapse
|
20
|
Small Molecule Effectors of Myosin Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1239:61-84. [DOI: 10.1007/978-3-030-38062-5_5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
21
|
Cota Teixeira S, Silva Lopes D, Santos da Silva M, Cordero da Luz FA, Cirilo Gimenes SN, Borges BC, Alves da Silva A, Alves Martins F, Alves Dos Santos M, Teixeira TL, Oliveira RA, de Melo Rodrigues Ávila V, Barbosa Silva MJ, Elias MC, Martin R, Vieira da Silva C, Knölker HJ. Pentachloropseudilin Impairs Angiogenesis by Disrupting the Actin Cytoskeleton, Integrin Trafficking and the Cell Cycle. Chembiochem 2019; 20:2390-2401. [PMID: 31026110 DOI: 10.1002/cbic.201900203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Indexed: 12/21/2022]
Abstract
Class 1 myosins (Myo1s) were the first unconventional myosins identified and humans have eight known Myo1 isoforms. The Myo1 family is involved in the regulation of gene expression, cytoskeletal rearrangements, delivery of proteins to the cell surface, cell migration and spreading. Thus, the important role of Myo1s in different biological processes is evident. In this study, we have investigated the effects of pentachloropseudilin (PClP), a reversible and allosteric potent inhibitor of Myo1s, on angiogenesis. We demonstrated that treatment of cells with PClP promoted a decrease in the number of vessels. The observed inhibition of angiogenesis is likely to be related to the inhibition of cell proliferation, migration and adhesion, as well as to alteration of the actin cytoskeleton pattern, as shown on a PClP-treated HUVEC cell line. Moreover, we also demonstrated that PClP treatment partially prevented the delivery of integrins to the plasma membrane. Finally, we showed that PClP caused DNA strand breaks, which are probably repaired during the cell cycle arrest in the G1 phase. Taken together, our results suggest that Myo1s participate directly in the angiogenesis process.
Collapse
Affiliation(s)
- Samuel Cota Teixeira
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Daiana Silva Lopes
- Multidisciplinary Institute of Health, Anísio Teixeira Campus, Federal University of Bahia, Rua Hormindo Barros, 58, Candeias, Vitória da Conquista, 45029-094, BA, Brazil
| | - Marcelo Santos da Silva
- Special Laboratory of Cell Cycle (LECC), Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, Av. Vital Brasil, 1500 - Butantã, São Paulo, 05503-900, SP, Brazil.,The Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, Sir Graeme Davies Building, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK
| | - Felipe Andrés Cordero da Luz
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Sarah Natalie Cirilo Gimenes
- Imunopathology Laboratory, Butantan Institute, Av. Vital Brasil, 1500 - Butantã, São Paulo, 05503-900, SP, Brazil
| | - Bruna Cristina Borges
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Aline Alves da Silva
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Flávia Alves Martins
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Marlus Alves Dos Santos
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Thaise Lara Teixeira
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Ricardo A Oliveira
- Medical School, Federal University of Uberlândia, Av. Pará, Bloco 2u, 1720 - Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Veridiana de Melo Rodrigues Ávila
- Institute of Biotechnology, Federal University of Uberlândia, Av. Pará, 1720 - Bloco 2E - Sala(s) 246 - Campus Umuarama, Uberlândia, 38405-320, MG, Brazil
| | - Marcelo José Barbosa Silva
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Maria Carolina Elias
- Special Laboratory of Cell Cycle (LECC), Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, Av. Vital Brasil, 1500 - Butantã, São Paulo, 05503-900, SP, Brazil
| | - René Martin
- Fakultät Chemie, Technische Universität Dresden, Bergstraße 66, 01069, Dresden, Germany
| | - Claudio Vieira da Silva
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Hans-Joachim Knölker
- Fakultät Chemie, Technische Universität Dresden, Bergstraße 66, 01069, Dresden, Germany
| |
Collapse
|
22
|
Kumar M, Sharma S, Sil P, Kushwaha M, Mayor S, Vishwakarma RA, Singh PP. C-H Arylation of N
-Heteroarenes under Metal-Free Conditions and its Application towards the Synthesis of Pentabromo- and Pentachloropseudilins. European J Org Chem 2019. [DOI: 10.1002/ejoc.201900353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Mukesh Kumar
- Medicinal Chemistry Division; Academy of Scientific and Innovative Research; CSIR-Indian Institute of Integrative Medicine; Canal Road -180001 Jammu India
| | - Shweta Sharma
- Medicinal Chemistry Division; Academy of Scientific and Innovative Research; CSIR-Indian Institute of Integrative Medicine; Canal Road -180001 Jammu India
| | - Parijat Sil
- National Centre for Biological Sciences; Tata Institute of Fundamental Research; GKVK; Bellary Road 560065 Bangalore- India
| | - Manoj Kushwaha
- Medicinal Chemistry Division; Academy of Scientific and Innovative Research; CSIR-Indian Institute of Integrative Medicine; Canal Road -180001 Jammu India
| | - Satyajit Mayor
- National Centre for Biological Sciences; Tata Institute of Fundamental Research; GKVK; Bellary Road 560065 Bangalore- India
| | - Ram A. Vishwakarma
- Medicinal Chemistry Division; Academy of Scientific and Innovative Research; CSIR-Indian Institute of Integrative Medicine; Canal Road -180001 Jammu India
| | - Parvinder Pal Singh
- Medicinal Chemistry Division; Academy of Scientific and Innovative Research; CSIR-Indian Institute of Integrative Medicine; Canal Road -180001 Jammu India
| |
Collapse
|
23
|
Wollenberg RD, Taft MH, Giese S, Thiel C, Balázs Z, Giese H, Manstein DJ, Sondergaard TE. Phenamacril is a reversible and noncompetitive inhibitor of Fusarium class I myosin. J Biol Chem 2019; 294:1328-1337. [PMID: 30504222 PMCID: PMC6349130 DOI: 10.1074/jbc.ra118.005408] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/01/2018] [Indexed: 01/03/2023] Open
Abstract
The cyanoacrylate compound phenamacril (also known as JS399-19) is a recently identified fungicide that exerts its antifungal effect on susceptible Fusarium species by inhibiting the ATPase activity of their myosin class I motor domains. Although much is known about the antifungal spectrum of phenamacril, the exact mechanism behind the phenamacril-mediated inhibition remains to be resolved. Here, we describe the characterization of the effect of phenamacril on purified myosin motor constructs from the model plant pathogen and phenamacril-susceptible species Fusarium graminearum, phenamacril-resistant Fusarium species, and the mycetozoan model organism Dictyostelium discoideum Our results show that phenamacril potently (IC50 ∼360 nm), reversibly, and noncompetitively inhibits ATP turnover, actin binding during ATP turnover, and motor activity of F. graminearum myosin-1. Phenamacril also inhibits the ATPase activity of Fusarium avenaceum myosin-1 but has little or no inhibitory effect on the motor activity of Fusarium solani myosin-1, human myosin-1c, and D. discoideum myosin isoforms 1B, 1E, and 2. Our findings indicate that phenamacril is a species-specific, noncompetitive inhibitor of class I myosin in susceptible Fusarium sp.
Collapse
Affiliation(s)
- Rasmus D Wollenberg
- Department of Chemistry and Bioscience, Aalborg University, DK-9220 Aalborg, Denmark
| | - Manuel H Taft
- Institute for Biophysical Chemistry, OE4350, Hannover Medical School, 30623 Hannover, Germany
| | - Sven Giese
- Institute for Biophysical Chemistry, OE4350, Hannover Medical School, 30623 Hannover, Germany
| | - Claudia Thiel
- Division of Structural Biochemistry, OE8830, Hannover Medical School, 30623 Hannover, Germany
| | - Zoltán Balázs
- Department of Chemistry and Bioscience, Aalborg University, DK-9220 Aalborg, Denmark
| | - Henriette Giese
- Department of Chemistry and Bioscience, Aalborg University, DK-9220 Aalborg, Denmark
| | - Dietmar J Manstein
- Institute for Biophysical Chemistry, OE4350, Hannover Medical School, 30623 Hannover, Germany; Division of Structural Biochemistry, OE8830, Hannover Medical School, 30623 Hannover, Germany.
| | - Teis E Sondergaard
- Department of Chemistry and Bioscience, Aalborg University, DK-9220 Aalborg, Denmark.
| |
Collapse
|
24
|
Structural and mechanistic insights into the function of the unconventional class XIV myosin MyoA from Toxoplasma gondii. Proc Natl Acad Sci U S A 2018; 115:E10548-E10555. [PMID: 30348763 DOI: 10.1073/pnas.1811167115] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Parasites of the phylum Apicomplexa are responsible for significant morbidity and mortality on a global scale. Central to the virulence of these pathogens are the phylum-specific, unconventional class XIV myosins that power the essential processes of parasite motility and host cell invasion. Notably, class XIV myosins differ from human myosins in key functional regions, yet they are capable of fast movement along actin filaments with kinetics rivaling previously studied myosins. Toward establishing a detailed molecular mechanism of class XIV motility, we determined the 2.6-Å resolution crystal structure of the Toxoplasma gondii MyoA (TgMyoA) motor domain. Structural analysis reveals intriguing strategies for force transduction and chemomechanical coupling that rely on a divergent SH1/SH2 region, the class-defining "HYAG"-site polymorphism, and the actin-binding surface. In vitro motility assays and hydrogen-deuterium exchange coupled with MS further reveal the mechanistic underpinnings of phosphorylation-dependent modulation of TgMyoA motility whereby localized regions of increased stability and order correlate with enhanced motility. Analysis of solvent-accessible pockets reveals striking differences between apicomplexan class XIV and human myosins. Extending these analyses to high-confidence homology models of Plasmodium and Cryptosporidium MyoA motor domains supports the intriguing potential of designing class-specific, yet broadly active, apicomplexan myosin inhibitors. The successful expression of the functional TgMyoA complex combined with our crystal structure of the motor domain provides a strong foundation in support of detailed structure-function studies and enables the development of small-molecule inhibitors targeting these devastating global pathogens.
Collapse
|
25
|
Iuliano O, Yoshimura A, Prospéri MT, Martin R, Knölker HJ, Coudrier E. Myosin 1b promotes axon formation by regulating actin wave propagation and growth cone dynamics. J Cell Biol 2018; 217:2033-2046. [PMID: 29588377 PMCID: PMC5987710 DOI: 10.1083/jcb.201703205] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 12/20/2017] [Accepted: 03/02/2018] [Indexed: 02/06/2023] Open
Abstract
Single-headed myosin 1 has been identified in neurons, but its function in these cells is still unclear. We demonstrate that depletion of myosin 1b (Myo1b), inhibition of its motor activity, or its binding to phosphoinositides impairs the formation of the axon, whereas overexpression of Myo1b increases the number of axon-like structures. Myo1b is associated with growth cones and actin waves, two major contributors to neuronal symmetry breaking. We show that Myo1b controls the dynamics of the growth cones and the anterograde propagation of the actin waves. By coupling the membrane to the actin cytoskeleton, Myo1b regulates the size of the actin network as well as the stability and size of filopodia in the growth cones. Our data provide the first evidence that a myosin 1 plays a major role in neuronal symmetry breaking and argue for a mechanical control of the actin cytoskeleton both in actin waves and in the growth cones by this myosin.
Collapse
Affiliation(s)
- Olga Iuliano
- Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherche Scientifique, UMR 144, Paris, France
| | - Azumi Yoshimura
- Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherche Scientifique, UMR 144, Paris, France
| | - Marie-Thérèse Prospéri
- Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherche Scientifique, UMR 144, Paris, France
| | - René Martin
- Department of Chemistry, Technische Univesität Dresden, Dresden, Germany
| | | | - Evelyne Coudrier
- Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherche Scientifique, UMR 144, Paris, France
| |
Collapse
|
26
|
Chung CL, Wang SW, Martin R, Knölker HJ, Kao YC, Lin MH, Chen JJ, Huang YB, Wu DC, Chen CL. Pentachloropseudilin Inhibits Transforming Growth Factor-β (TGF-β) Activity by Accelerating Cell-Surface Type II TGF-β Receptor Turnover in Target Cells. Chembiochem 2018; 19:851-864. [DOI: 10.1002/cbic.201700693] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Chih-Ling Chung
- Department of Biological Sciences; National Sun Yat-sen University; Kaohsiung 80424 ROC Taiwan
| | - Shih-Wei Wang
- Department of Biological Sciences; National Sun Yat-sen University; Kaohsiung 80424 ROC Taiwan
| | - René Martin
- Department of Chemistry; Technische Universität Dresden; Bergstrasse 66 01069 Dresden Germany
| | - Hans-Joachim Knölker
- Department of Chemistry; Technische Universität Dresden; Bergstrasse 66 01069 Dresden Germany
| | - Yu-Chen Kao
- Department of Biological Sciences; National Sun Yat-sen University; Kaohsiung 80424 ROC Taiwan
| | - Ming-Hong Lin
- Department of Microbiology and Immunology; Faculty of Medicine; Kaohsiung Medical University Hospital; Kaohsiung 80708 ROC Taiwan
| | - Jih-Jung Chen
- Faculty of Pharmacy; School of Pharmaceutical Sciences; National Yang-Ming University; Taipei 11221 ROC Taiwan
| | - Yaw-Bin Huang
- Department of Biological Sciences; National Sun Yat-sen University; Kaohsiung 80424 ROC Taiwan
- Department of Pharmacy; School of Pharmacy; Kaohsiung Medical University; Kaohsiung 80708 ROC Taiwan
- Center for Stem Cell Research; Kaohsiung Medical University; Kaohsiung 80708 ROC Taiwan
| | - Deng-Chyang Wu
- Division of Gastroenterology; Department of Internal Medicine; Kaohsiung Medical University Hospital; Kaohsiung 80708 ROC Taiwan
- Center for Stem Cell Research; Kaohsiung Medical University; Kaohsiung 80708 ROC Taiwan
| | - Chun-Lin Chen
- Department of Biological Sciences; National Sun Yat-sen University; Kaohsiung 80424 ROC Taiwan
- Doctoral Degree Program in Marine Biotechnology; National Sun Yat-sen University and Academia Sinica; Kaohsiung 80424 ROC Taiwan
| |
Collapse
|
27
|
Vallotton P, van Oijen AM, Whitchurch CB, Gelfand V, Yeo L, Tsiavaliaris G, Heinrich S, Dultz E, Weis K, Grünwald D. Diatrack particle tracking software: Review of applications and performance evaluation. Traffic 2017; 18:840-852. [PMID: 28945316 PMCID: PMC5677553 DOI: 10.1111/tra.12530] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 09/21/2017] [Accepted: 09/21/2017] [Indexed: 12/24/2022]
Abstract
Object tracking is an instrumental tool supporting studies of cellular trafficking. There are three challenges in object tracking: the identification of targets; the precise determination of their position and boundaries; and the assembly of correct trajectories. This last challenge is particularly relevant when dealing with densely populated images with low signal-to-noise ratios-conditions that are often encountered in applications such as organelle tracking, virus particle tracking or single-molecule imaging. We have developed a set of methods that can handle a wide variety of signal complexities. They are compiled into a free software package called Diatrack. Here we review its main features and utility in a range of applications, providing a survey of the dynamic imaging field together with recommendations for effective use. The performance of our framework is shown to compare favorably to a wide selection of custom-developed algorithms, whether in terms of localization precision, processing speed or correctness of tracks.
Collapse
Affiliation(s)
| | | | | | - Vladimir Gelfand
- Northwestern University Feinberg School of Medicine, Department of Cell and Molecular Biology, Chicago, IL 60611, USA
| | | | | | | | - Elisa Dultz
- ETH Zürich, Institute of Biochemistry, Zürich, Switzerland
| | - Karsten Weis
- ETH Zürich, Institute of Biochemistry, Zürich, Switzerland
| | - David Grünwald
- University of Massachusetts Medical School, RNA Therapeutics Institute and Department of Biochemistry and Molecular Pharmacology, Worcester MA, USA
| |
Collapse
|
28
|
The Conserved Lysine-265 Allosterically Modulates Nucleotide- and Actin-binding Site Coupling in Myosin-2. Sci Rep 2017; 7:7650. [PMID: 28794442 PMCID: PMC5550493 DOI: 10.1038/s41598-017-07933-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/05/2017] [Indexed: 11/18/2022] Open
Abstract
Myosin motor proteins convert chemical energy into force and movement through their interactions with nucleotide and filamentous actin (F-actin). The evolutionarily conserved lysine-265 (K265) of the myosin-2 motor from Dictyostelium discoideum (Dd) is proposed to be a key residue in an allosteric communication pathway that mediates actin-nucleotide coupling. To better understand the role of K265, point mutations were introduced within the Dd myosin-2 M765-2R framework, replacing this lysine with alanine (K265A), glutamic acid (K265E) or glutamine (K265Q), and the functional and kinetic properties of the resulting myosin motors were assessed. The alanine and glutamic acid substitutions reduced actin-activated ATPase activity, slowed the in vitro sliding velocity and attenuated the inhibitory potential of the allosteric myosin inhibitor pentabromopseudilin (PBP). However, glutamine substitution did not substantially change these parameters. Structural modelling suggests that K265 interacts with D590 and Q633 to establish a pivotal allosteric branching point. Based on our results, we propose: (1) that the K265-D590 interaction functions to reduce myosins basal ATPase activity in the absence of F-actin, and (2) that the dynamic formation of the K265-Q633 salt bridge upon actin cleft closure regulates the activation of product release by actin filaments.
Collapse
|
29
|
Gupta P, Martin R, Knölker HJ, Nihalani D, Kumar Sinha D. Myosin-1 inhibition by PClP affects membrane shape, cortical actin distribution and lipid droplet dynamics in early Zebrafish embryos. PLoS One 2017; 12:e0180301. [PMID: 28678859 PMCID: PMC5498032 DOI: 10.1371/journal.pone.0180301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/13/2017] [Indexed: 12/22/2022] Open
Abstract
Myosin-1 (Myo1) represents a mechanical link between the membrane and actin-cytoskeleton in animal cells. We have studied the effect of Myo1 inhibitor PClP in 1-8 cell Zebrafish embryos. Our results indicate a unique involvement of Myo1 in early development of Zebrafish embryos. Inhibition of Myo1 (by PClP) and Myo2 (by Blebbistatin) lead to arrest in cell division. While Myo1 isoforms appears to be important for both the formation and the maintenance of cleavage furrows, Myo2 is required only for the formation of furrows. We found that the blastodisc of the embryo, which contains a thick actin cortex (~13 μm), is loaded with cortical Myo1. Myo1 appears to be crucial for maintaining the blastodisc morphology and the actin cortex thickness. In addition to cell division and furrow formation, inhibition of Myo1 has a drastic effect on the dynamics and distribution of lipid droplets (LDs) in the blastodisc near the cleavage furrow. All these results above are effects of Myo1 inhibition exclusively; Myo2 inhibition by blebbistatin does not show such phenotypes. Therefore, our results demonstrate a potential role for Myo1 in the maintenance and formation of furrow, blastodisc morphology, cell-division and LD organization within the blastodisc during early embryogenesis.
Collapse
MESH Headings
- Actin Cytoskeleton/drug effects
- Actin Cytoskeleton/metabolism
- Actins/genetics
- Actins/metabolism
- Animals
- Blastomeres/cytology
- Blastomeres/metabolism
- Blastomeres/ultrastructure
- Blotting, Western
- Cell Division/drug effects
- Cell Division/genetics
- Cell Membrane/metabolism
- Embryo, Nonmammalian/embryology
- Embryo, Nonmammalian/metabolism
- Embryo, Nonmammalian/ultrastructure
- Gene Expression Regulation, Developmental
- Heterocyclic Compounds, 4 or More Rings/pharmacology
- Hydrocarbons, Chlorinated/pharmacology
- Lipid Droplets/metabolism
- Microscopy, Electron, Scanning
- Microscopy, Fluorescence
- Myosin Heavy Chains/antagonists & inhibitors
- Myosin Heavy Chains/genetics
- Myosin Heavy Chains/metabolism
- Pyrroles/pharmacology
- Reverse Transcriptase Polymerase Chain Reaction
- Zebrafish/embryology
- Zebrafish/genetics
- Zebrafish/metabolism
- Zebrafish Proteins/antagonists & inhibitors
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
| | - René Martin
- Department Chemie, TU Dresden, Dresden, Germany
| | | | - Deepak Nihalani
- Dept. Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | | |
Collapse
|
30
|
Masters TA, Kendrick-Jones J, Buss F. Myosins: Domain Organisation, Motor Properties, Physiological Roles and Cellular Functions. Handb Exp Pharmacol 2017; 235:77-122. [PMID: 27757761 DOI: 10.1007/164_2016_29] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Myosins are cytoskeletal motor proteins that use energy derived from ATP hydrolysis to generate force and movement along actin filaments. Humans express 38 myosin genes belonging to 12 classes that participate in a diverse range of crucial activities, including muscle contraction, intracellular trafficking, cell division, motility, actin cytoskeletal organisation and cell signalling. Myosin malfunction has been implicated a variety of disorders including deafness, hypertrophic cardiomyopathy, Usher syndrome, Griscelli syndrome and cancer. In this chapter, we will first discuss the key structural and kinetic features that are conserved across the myosin family. Thereafter, we summarise for each member in turn its unique functional and structural adaptations, cellular roles and associated pathologies. Finally, we address the broad therapeutic potential for pharmacological interventions that target myosin family members.
Collapse
Affiliation(s)
- Thomas A Masters
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK.
| | | | - Folma Buss
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| |
Collapse
|
31
|
Wollenberg RD, Donau SS, Nielsen TT, Sørensen JL, Giese H, Wimmer R, Søndergaard TE. Real-time imaging of the growth-inhibitory effect of JS399-19 on Fusarium. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2016; 134:24-30. [PMID: 27914536 DOI: 10.1016/j.pestbp.2016.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/03/2016] [Accepted: 05/09/2016] [Indexed: 06/06/2023]
Abstract
Real-time imaging was used to study the effects of a novel Fusarium-specific cyanoacrylate fungicide (JS399-19) on growth and morphology of four Fusarium sp. This fungicide targets the motor domain of type I myosin. Fusarium graminearum PH-1, Fusarium solani f. sp. pisi 77-13-4, Fusarium avenaceum IBT8464, and Fusarium avenaceum 05001, which has a K216Q amino-acid substitution at the resistance-implicated site in its myosin type I motor domain, were analyzed. Real-time imaging shows that JS399-19 inhibits fungal growth but not to the extent previously reported. The fungicide causes the hypha to become entangled and unable to extend vertically. This implies that type I myosin in Fusarium is essential for hyphal and mycelia propagation. The K216Q substitution correlates with reduced susceptibility in F. avenaceum.
Collapse
Affiliation(s)
- Rasmus D Wollenberg
- Department of Chemistry and Bioscience, Aalborg University, DK-9220 Aalborg, Denmark.
| | - Søren S Donau
- Department of Chemistry and Bioscience, Aalborg University, DK-9220 Aalborg, Denmark
| | - Thorbjørn T Nielsen
- Department of Chemistry and Bioscience, Aalborg University, DK-9220 Aalborg, Denmark
| | - Jens L Sørensen
- Department of Chemistry and Bioscience, Aalborg University, DK-9220 Aalborg, Denmark
| | - Henriette Giese
- Department of Chemistry and Bioscience, Aalborg University, DK-9220 Aalborg, Denmark
| | - Reinhard Wimmer
- Department of Chemistry and Bioscience, Aalborg University, DK-9220 Aalborg, Denmark
| | - Teis E Søndergaard
- Department of Chemistry and Bioscience, Aalborg University, DK-9220 Aalborg, Denmark
| |
Collapse
|
32
|
Prospéri MT, Lépine P, Dingli F, Paul-Gilloteaux P, Martin R, Loew D, Knölker HJ, Coudrier E. Myosin 1b functions as an effector of EphB signaling to control cell repulsion. J Cell Biol 2016. [PMID: 26195670 PMCID: PMC4508888 DOI: 10.1083/jcb.201501018] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Myosin 1b functions as an effector of EphB2/ephrinB signaling and controls cell morphology and cell repulsion. Eph receptors and their membrane-tethered ligands, the ephrins, have important functions in embryo morphogenesis and in adult tissue homeostasis. Eph/ephrin signaling is essential for cell segregation and cell repulsion. This process is accompanied by morphological changes and actin remodeling that drives cell segregation and tissue patterning. The actin cortex must be mechanically coupled to the plasma membrane to orchestrate the cell morphology changes. Here, we demonstrate that myosin 1b that can mechanically link the membrane to the actin cytoskeleton interacts with EphB2 receptors via its tail and is tyrosine phosphorylated on its tail in an EphB2-dependent manner. Myosin 1b regulates the redistribution of myosin II in actomyosin fibers and the formation of filopodia at the interface of ephrinB1 and EphB2 cells, which are two processes mediated by EphB2 signaling that contribute to cell repulsion. Together, our results provide the first evidence that a myosin 1 functions as an effector of EphB2/ephrinB signaling, controls cell morphology, and thereby cell repulsion.
Collapse
Affiliation(s)
| | - Priscilla Lépine
- Institut Curie, Centre de Recherche, F-75248 Paris, France Université Pierre et Marie Curie, F-75252 Paris, France
| | - Florent Dingli
- Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, Centre de Recherche, F-75248 Paris, France
| | - Perrine Paul-Gilloteaux
- Institut Curie, Centre de Recherche, F-75248 Paris, France Cell and Tissue Imaging Facility (PICT-IBiSA), Centre National de la Recherche Scientifique, UMR 144, Paris F-75248, France
| | - René Martin
- Department of Chemistry, Technische Univesität, 01069 Dresden, Germany
| | - Damarys Loew
- Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, Centre de Recherche, F-75248 Paris, France
| | | | - Evelyne Coudrier
- Institut Curie, Centre de Recherche, F-75248 Paris, France Cell and Tissue Imaging Facility (PICT-IBiSA), Centre National de la Recherche Scientifique, UMR 144, Paris F-75248, France
| |
Collapse
|
33
|
Kittelberger N, Breunig M, Martin R, Knölker HJ, Miklavc P. The role of myosin 1c and myosin 1b in surfactant exocytosis. J Cell Sci 2016; 129:1685-96. [PMID: 26940917 PMCID: PMC4852769 DOI: 10.1242/jcs.181313] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 02/25/2016] [Indexed: 12/19/2022] Open
Abstract
Actin and actin-associated proteins have a pivotal effect on regulated exocytosis in secretory cells and influence pre-fusion as well as post-fusion stages of exocytosis. Actin polymerization on secretory granules during the post-fusion phase (formation of an actin coat) is especially important in cells with large secretory vesicles or poorly soluble secretions. Alveolar type II (ATII) cells secrete hydrophobic lipo-protein surfactant, which does not easily diffuse from fused vesicles. Previous work showed that compression of actin coat is necessary for surfactant extrusion. Here, we investigate the role of class 1 myosins as possible linkers between actin and membranes during exocytosis. Live-cell microscopy showed translocation of fluorescently labeled myosin 1b and myosin 1c to the secretory vesicle membrane after fusion. Myosin 1c translocation was dependent on its pleckstrin homology domain. Expression of myosin 1b and myosin 1c constructs influenced vesicle compression rate, whereas only the inhibition of myosin 1c reduced exocytosis. These findings suggest that class 1 myosins participate in several stages of ATII cell exocytosis and link actin coats to the secretory vesicle membrane to influence vesicle compression.
Collapse
Affiliation(s)
- Nadine Kittelberger
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, Ulm 89081, Germany
| | - Markus Breunig
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, Ulm 89081, Germany
| | - René Martin
- Department of Chemistry, Technische Universität Dresden, Bergstr. 66, Dresden 01069, Germany
| | - Hans-Joachim Knölker
- Department of Chemistry, Technische Universität Dresden, Bergstr. 66, Dresden 01069, Germany
| | - Pika Miklavc
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, Ulm 89081, Germany
| |
Collapse
|
34
|
Heissler SM, Sellers JR. Various Themes of Myosin Regulation. J Mol Biol 2016; 428:1927-46. [PMID: 26827725 DOI: 10.1016/j.jmb.2016.01.022] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/12/2016] [Accepted: 01/19/2016] [Indexed: 10/24/2022]
Abstract
Members of the myosin superfamily are actin-based molecular motors that are indispensable for cellular homeostasis. The vast functional and structural diversity of myosins accounts for the variety and complexity of the underlying allosteric regulatory mechanisms that determine the activation or inhibition of myosin motor activity and enable precise timing and spatial aspects of myosin function at the cellular level. This review focuses on the molecular basis of posttranslational regulation of eukaryotic myosins from different classes across species by allosteric intrinsic and extrinsic effectors. First, we highlight the impact of heavy and light chain phosphorylation. Second, we outline intramolecular regulatory mechanisms such as autoinhibition and subsequent activation. Third, we discuss diverse extramolecular allosteric mechanisms ranging from actin-linked regulatory mechanisms to myosin:cargo interactions. At last, we briefly outline the allosteric regulation of myosins with synthetic compounds.
Collapse
Affiliation(s)
- Sarah M Heissler
- Laboratory of Molecular Physiology, National Heart, Lung and Blood Institute, National Institutes of Health, 50 South Drive, B50/3529, Bethesda, MD 20892-8015, USA.
| | - James R Sellers
- Laboratory of Molecular Physiology, National Heart, Lung and Blood Institute, National Institutes of Health, 50 South Drive, B50/3529, Bethesda, MD 20892-8015, USA
| |
Collapse
|
35
|
Brandstaetter H, Kishi-Itakura C, Tumbarello DA, Manstein DJ, Buss F. Loss of functional MYO1C/myosin 1c, a motor protein involved in lipid raft trafficking, disrupts autophagosome-lysosome fusion. Autophagy 2015; 10:2310-23. [PMID: 25551774 PMCID: PMC4502697 DOI: 10.4161/15548627.2014.984272] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
MYO1C, a single-headed class I myosin, associates with cholesterol-enriched lipid rafts and facilitates their recycling from intracellular compartments to the cell surface. Absence of functional MYO1C disturbs the cellular distribution of lipid rafts, causes the accumulation of cholesterol-enriched membranes in the perinuclear recycling compartment, and leads to enlargement of endolysosomal membranes. Several feeder pathways, including classical endocytosis but also the autophagy pathway, maintain the health of the cell by selective degradation of cargo through fusion with the lysosome. Here we show that loss of functional MYO1C leads to an increase in total cellular cholesterol and its disrupted subcellular distribution. We observe an accumulation of autophagic structures caused by a block in fusion with the lysosome and a defect in autophagic cargo degradation. Interestingly, the loss of MYO1C has no effect on degradation of endocytic cargo such as EGFR, illustrating that although the endolysosomal compartment is enlarged in size, it is functional, contains active hydrolases, and the correct pH. Our results highlight the importance of correct lipid composition in autophagosomes and lysosomes to enable them to fuse. Ablating MYO1C function causes abnormal cholesterol distribution, which has a major selective impact on the autophagy pathway.
Collapse
Key Words
- BafA1, bafilomycin A1
- EGF, epidermal growth factor
- EGFR, epidermal growth factor receptor
- EM, electron microscopy
- GFP, green fluorescent protein
- KD, knockdown
- LAMP1, lysosomal-associated membrane protein 1
- LC3, microtubule-associated protein 1 light chain 3
- MVB, multivesicular body
- MYO1C, myosin IC
- PB, phosphate buffer
- PCIP, pentachloropseudilin
- PtdIns(4, 5)P2, phosphatidylinositol 4, 5-bisphosphate
- RFP, red fluorescent protein
- RPE, retinal pigment epithelium
- autophagy
- cholesterol
- electron microscopy
- lipid raft
- lysosome, MYO1C
Collapse
Affiliation(s)
- Hemma Brandstaetter
- a Cambridge Institute for Medical Research ; University of Cambridge ; Cambridge , UK
| | | | | | | | | |
Collapse
|
36
|
Costa Pessoa J, Garribba E, Santos MF, Santos-Silva T. Vanadium and proteins: Uptake, transport, structure, activity and function. Coord Chem Rev 2015. [DOI: 10.1016/j.ccr.2015.03.016] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
37
|
Myosin-II-mediated cell shape changes and cell intercalation contribute to primitive streak formation. Nat Cell Biol 2015; 17:397-408. [PMID: 25812521 PMCID: PMC4886837 DOI: 10.1038/ncb3138] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 02/16/2015] [Indexed: 12/14/2022]
Abstract
Primitive streak formation in the chick embryo involves large scale highly coordinated flows of over 100.000 cells in the epiblast. These large scale tissue flows and deformations can be correlated with specific anisotropic cell behaviours in the forming mesendoderm through a combined light-sheet microscopy and computational analysis. Relevant behaviours include apical contraction, elongation along the apical-basal axis followed by ingression as well as asynchronous directional cell intercalation of small groups of mesendoderm cells. Cell intercalation is associated with sequential, directional contraction of apical junctions, the onset, localisation and direction of which correlate strongly with the appearance of active Myosin II cables in aligned apical junctions in neighbouring cells. Use of a class specific Myosin inhibitors and gene specific knockdowns show that apical contraction and intercalation are Myosin II dependent and also reveal critical roles for Myosin I and Myosin V family members in the assembly of junctional Myosin II cables.
Collapse
|
38
|
Poorly understood aspects of striated muscle contraction. BIOMED RESEARCH INTERNATIONAL 2015; 2015:245154. [PMID: 25961006 PMCID: PMC4415482 DOI: 10.1155/2015/245154] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 10/28/2014] [Indexed: 11/23/2022]
Abstract
Muscle contraction results from cyclic interactions between the contractile proteins myosin and actin, driven by the turnover of adenosine triphosphate (ATP). Despite intense studies, several molecular events in the contraction process are poorly understood, including the relationship between force-generation and phosphate-release in the ATP-turnover. Different aspects of the force-generating transition are reflected in the changes in tension development by muscle cells, myofibrils and single molecules upon changes in temperature, altered phosphate concentration, or length perturbations. It has been notoriously difficult to explain all these events within a given theoretical framework and to unequivocally correlate observed events with the atomic structures of the myosin motor. Other incompletely understood issues include the role of the two heads of myosin II and structural changes in the actin filaments as well as the importance of the three-dimensional order. We here review these issues in relation to controversies regarding basic physiological properties of striated muscle. We also briefly consider actomyosin mutation effects in cardiac and skeletal muscle function and the possibility to treat these defects by drugs.
Collapse
|
39
|
Zhang C, Chen Y, Yin Y, Ji HH, Shim WB, Hou Y, Zhou M, Li XD, Ma Z. A small molecule species specifically inhibits Fusarium myosin I. Environ Microbiol 2015; 17:2735-46. [PMID: 25404531 DOI: 10.1111/1462-2920.12711] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 11/03/2014] [Accepted: 11/05/2014] [Indexed: 11/28/2022]
Abstract
Fusarium head blight (FHB) caused by Fusarium graminearum is a devastating disease of cereal crops worldwide. Recently, a novel fungicide JS399-19 has been launched into the marketplace to manage FHB. It is compelling that JS399-19 shows highly inhibitory activity towards some Fusarium species, but not to other fungi, indicating that it is an environmentally compatible fungicide. To explore the mode of action of this species-specific compound, we conducted a whole-genome transcript profiling together with genetic and biochemical assays, and discovered that JS399-19 targets the myosin I of F. graminearum (FgMyo1). FgMyo1 is essential for F. graminearum growth. A point mutation S217L or E420K in FgMyo1 is responsible for F. graminearum resistance to JS399-19. In addition, transformation of F. graminearum with the myosin I gene of Magnaporthe grisea, the causal agent of rice blast, also led to JS399-19 resistance. JS399-19 strongly inhibits the ATPase activity of the wild-type FgMyo1, but not the mutated FgMyo1(S217L/E420K) . These results provide us a new insight into the design of species-specific antifungal compounds. Furthermore, our strategy can be applied to identify novel drug targets in various pathogenic organisms.
Collapse
Affiliation(s)
- Chengqi Zhang
- Institute of Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Yun Chen
- Institute of Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Yanni Yin
- Institute of Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Huan-Hong Ji
- National Laboratory of Integrated Management of Insect Pests and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Won-Bo Shim
- Department of Plant Pathology and Microbiology, Texas A&M University, College Station, TX, 77843-2132, USA
| | - Yiping Hou
- Department of Plant Pathology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Mingguo Zhou
- Department of Plant Pathology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiang-Dong Li
- National Laboratory of Integrated Management of Insect Pests and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhonghua Ma
- Institute of Biotechnology, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
40
|
Ohmura G, Tsujikawa T, Yaguchi T, Kawamura N, Mikami S, Sugiyama J, Nakamura K, Kobayashi A, Iwata T, Nakano H, Shimada T, Hisa Y, Kawakami Y. Aberrant Myosin 1b Expression Promotes Cell Migration and Lymph Node Metastasis of HNSCC. Mol Cancer Res 2014; 13:721-31. [PMID: 25421751 DOI: 10.1158/1541-7786.mcr-14-0410] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 11/12/2014] [Indexed: 11/16/2022]
Abstract
UNLABELLED Lymph node metastasis is the major clinicopathologic feature associated with poor prognosis in patients with head and neck squamous cell carcinoma (HNSCC). Here, web-based bioinformatics meta-analysis was performed to elucidate the molecular mechanism of lymph node metastasis of human HNSCC. Preferential upregulation of Myosin 1b (MYO1B) transcript in HNSCC datasets was identified. Myo1b mRNA was highly expressed in human HNSCC cells and patient tissue specimens compared with their normal counterparts as shown by quantitative PCR (qPCR) analyses. Immunohistochemistry (IHC)-detected Myo1b expression was significantly correlated with lymph node metastases in patients with oral cancer of the tongue. HNSCC with high expression of Myo1b and chemokine receptor 4 (CCR4), another metastasis-associated molecule, was strongly associated with lymph node metastasis. RNA interference (RNAi) of Myo1b in HNSCC cells, SAS and HSC4, significantly inhibited migratory and invasive abilities through decreased large protrusion formation of cell membranes. Finally, Myo1b knockdown in SAS cells significantly inhibited in vivo cervical lymph node metastases in a cervical lymph node metastatic mouse model system. IMPLICATIONS Myo1b is functionally involved in lymph node metastasis of human HNSCC through enhanced cancer cell motility and is an attractive target for new diagnostic and therapeutic strategies for patients with HNSCC.
Collapse
Affiliation(s)
- Gaku Ohmura
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan. Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto City, Kyoto, Japan
| | - Takahiro Tsujikawa
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan. Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto City, Kyoto, Japan
| | - Tomonori Yaguchi
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Naoshi Kawamura
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Shuji Mikami
- Division of Diagnostic Pathology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Juri Sugiyama
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kenta Nakamura
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Asuka Kobayashi
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Takashi Iwata
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hiroshi Nakano
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto City, Kyoto, Japan
| | - Taketoshi Shimada
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto City, Kyoto, Japan
| | - Yasuo Hisa
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto City, Kyoto, Japan
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan.
| |
Collapse
|
41
|
Martin R, Risacher C, Barthel A, Jäger A, Schmidt AW, Richter S, Böhl M, Preller M, Chinthalapudi K, Manstein DJ, Gutzeit HO, Knölker HJ. Silver(I)-Catalyzed Route to Pyrroles: Synthesis of Halogenated Pseudilins as Allosteric Inhibitors for Myosin ATPase and X-ray Crystal Structures of the Protein-Inhibitor Complexes. European J Org Chem 2014. [DOI: 10.1002/ejoc.201402177] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
42
|
Arjonen A, Kaukonen R, Mattila E, Rouhi P, Högnäs G, Sihto H, Miller BW, Morton JP, Bucher E, Taimen P, Virtakoivu R, Cao Y, Sansom OJ, Joensuu H, Ivaska J. Mutant p53-associated myosin-X upregulation promotes breast cancer invasion and metastasis. J Clin Invest 2014; 124:1069-82. [PMID: 24487586 PMCID: PMC3934176 DOI: 10.1172/jci67280] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 11/14/2013] [Indexed: 02/04/2023] Open
Abstract
Mutations of the tumor suppressor TP53 are present in many forms of human cancer and are associated with increased tumor cell invasion and metastasis. Several mechanisms have been identified for promoting dissemination of cancer cells with TP53 mutations, including increased targeting of integrins to the plasma membrane. Here, we demonstrate a role for the filopodia-inducing motor protein Myosin-X (Myo10) in mutant p53-driven cancer invasion. Analysis of gene expression profiles from 2 breast cancer data sets revealed that MYO10 was highly expressed in aggressive cancer subtypes. Myo10 was required for breast cancer cell invasion and dissemination in multiple cancer cell lines and murine models of cancer metastasis. Evaluation of a Myo10 mutant without the integrin-binding domain revealed that the ability of Myo10 to transport β₁ integrins to the filopodia tip is required for invasion. Introduction of mutant p53 promoted Myo10 expression in cancer cells and pancreatic ductal adenocarcinoma in mice, whereas suppression of endogenous mutant p53 attenuated Myo10 levels and cell invasion. In clinical breast carcinomas, Myo10 was predominantly expressed at the invasive edges and correlated with the presence of TP53 mutations and poor prognosis. These data indicate that Myo10 upregulation in mutant p53-driven cancers is necessary for invasion and that plasma-membrane protrusions, such as filopodia, may serve as specialized metastatic engines.
Collapse
Affiliation(s)
- Antti Arjonen
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Riina Kaukonen
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Elina Mattila
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Pegah Rouhi
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Gunilla Högnäs
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Harri Sihto
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Bryan W. Miller
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Jennifer P. Morton
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Elmar Bucher
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Pekka Taimen
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Reetta Virtakoivu
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Yihai Cao
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Owen J. Sansom
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Heikki Joensuu
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Johanna Ivaska
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| |
Collapse
|
43
|
ten Siethoff L, Lard M, Generosi J, Andersson H, Linke H, Månsson A. Molecular motor propelled filaments reveal light-guiding in nanowire arrays for enhanced biosensing. NANO LETTERS 2014; 14:737-42. [PMID: 24367994 PMCID: PMC3924849 DOI: 10.1021/nl404032k] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/17/2013] [Indexed: 05/27/2023]
Abstract
Semiconductor nanowire arrays offer significant potential for biosensing applications with optical read-out due to their high surface area and due to the unique optical properties of one-dimensional materials. A challenge for optical read-out of analyte-binding to the nanowires is the need to efficiently collect and detect light from a three-dimensional volume. Here we show that light from fluorophores attached along several μm long vertical Al2O3 coated gallium phosphide nanowires couples into the wires, is guided along them and emitted at the tip. This enables effective collection of light emitted by fluorescent analytes located at different focal planes along the nanowire. We unequivocally demonstrate the light-guiding effect using a novel method whereby the changes in emitted fluorescence intensity are observed when fluorescent cytoskeletal filaments are propelled by molecular motors along the wires. The findings are discussed in relation to nanobiosensor developments, other nanotechnological applications, and fundamental studies of motor function.
Collapse
Affiliation(s)
- Lasse ten Siethoff
- Department
of Chemistry and Biomedical Sciences, Linnaeus
University, SE-391 82 Kalmar, Sweden
| | - Mercy Lard
- Nanometer
Structure Consortium (nmC@LU) and Solid State Physics Lund University, SE-221 00 Lund, Sweden
| | - Johanna Generosi
- Nanometer
Structure Consortium (nmC@LU) and Solid State Physics Lund University, SE-221 00 Lund, Sweden
| | - Håkan
S. Andersson
- Department
of Chemistry and Biomedical Sciences, Linnaeus
University, SE-391 82 Kalmar, Sweden
| | - Heiner Linke
- Nanometer
Structure Consortium (nmC@LU) and Solid State Physics Lund University, SE-221 00 Lund, Sweden
| | - Alf Månsson
- Department
of Chemistry and Biomedical Sciences, Linnaeus
University, SE-391 82 Kalmar, Sweden
| |
Collapse
|
44
|
Gupta P, Gauthier NC, Cheng-Han Y, Zuanning Y, Pontes B, Ohmstede M, Martin R, Knölker HJ, Döbereiner HG, Krendel M, Sheetz M. Myosin 1E localizes to actin polymerization sites in lamellipodia, affecting actin dynamics and adhesion formation. Biol Open 2013; 2:1288-99. [PMID: 24337113 PMCID: PMC3863413 DOI: 10.1242/bio.20135827] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Because the actin network in active lamellipodia is continuously assembling at the edge, moving inward and disassembling, there is a question as to how actin-binding proteins and other components are transported to the leading edge and how nascent adhesions are stabilized. Active transport could play a significant role in these functions but the components involved are unknown. We show here that Myosin 1E (a long tailed Myosin 1 isoform) rapidly moves to the tips of active lamellipodia and to actin-rich early adhesions, unlike Myosin 1G, 1B or 1C (short tailed isoforms). Myosin 1E co-localizes with CARMIL, FHOD1, Arp3 and β3-integrin in those early adhesions. But these structures precede stable paxillin-rich adhesions. Myosin 1E movement depends upon actin-binding domains and the presence of an SH3 oligomerization domain. Overexpression of a Myosin 1E deletion mutant without the extreme C-terminal interacting (SH3) domain (Myosin 1EΔSH3) increases edge fluctuations and decreases stable adhesion lifetimes. In contrast, overexpression of Myosin 1E full tail domain (TH1+TH2+TH3/SH3) decreases edge fluctuation. In Myosin 1E knockdown cells, and more prominently in cells treated with Myosin 1 inhibitor, cell-matrix adhesions are also short-lived and fail to mature. We suggest that, by moving to actin polymerization sites and early adhesion sites in active lamellipodia, Myosin 1E might play important roles in transporting not only important polymerizing proteins but also proteins involved in adhesion stabilization.
Collapse
Affiliation(s)
- Prabuddha Gupta
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Preller M, Manstein D. Myosin Structure, Allostery, and Mechano-Chemistry. Structure 2013; 21:1911-22. [DOI: 10.1016/j.str.2013.09.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 09/19/2013] [Accepted: 09/25/2013] [Indexed: 01/10/2023]
|
46
|
Bond LM, Tumbarello DA, Kendrick-Jones J, Buss F. Small-molecule inhibitors of myosin proteins. Future Med Chem 2013; 5:41-52. [PMID: 23256812 PMCID: PMC3971371 DOI: 10.4155/fmc.12.185] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Advances in screening and computational methods have enhanced recent efforts to discover/design small-molecule protein inhibitors. One attractive target for inhibition is the myosin family of motor proteins. Myosins function in a wide variety of cellular processes, from intracellular trafficking to cell motility, and are implicated in several human diseases (e.g., cancer, hypertrophic cardiomyopathy, deafness and many neurological disorders). Potent and selective myosin inhibitors are, therefore, not only a tool for understanding myosin function, but are also a resource for developing treatments for diseases involving myosin dysfunction or overactivity. This review will provide a brief overview of the characteristics and scientific/therapeutic applications of the presently identified small-molecule myosin inhibitors before discussing the future of myosin inhibitor and activator design.
Collapse
Affiliation(s)
- Lisa M Bond
- Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, UK
| | - David A Tumbarello
- Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, UK
| | | | - Folma Buss
- Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, UK
| |
Collapse
|
47
|
Bond LM, Brandstaetter H, Kendrick-Jones J, Buss F. Functional roles for myosin 1c in cellular signaling pathways. Cell Signal 2013; 25:229-35. [PMID: 23022959 PMCID: PMC3715701 DOI: 10.1016/j.cellsig.2012.09.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 09/24/2012] [Indexed: 02/01/2023]
Abstract
Cellular signaling pathways underlie the transfer of information throughout the cell and to adjoining cells and so govern most critical cellular functions. Increasing evidence points to the molecular motor myosin 1c as a prominent player in many signaling cascades, from the integrin-dependent signaling involved in cell migration to the signaling events underlying insulin resistance. Myosin 1c functions on these pathways both via an important role in regulating lipid raft recycling and also via direct involvement in signaling cascades. This review provides an overview of the functional involvement of myosin 1c in cellular signaling and discusses the possible potential for myosin 1c as a target for drug-based treatments for human diseases.
Collapse
Affiliation(s)
- Lisa M Bond
- Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, United Kingdom
| | | | | | | |
Collapse
|
48
|
van Zalinge H, Aveyard J, Hajne J, Persson M, Mansson A, Nicolau DV. Actin filament motility induced variation of resonance frequency and rigidity of polymer surfaces studied by quartz crystal microbalance. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2012; 28:15033-15037. [PMID: 22988957 DOI: 10.1021/la302717y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
This contribution reports on the quantification of the parameters of the motility assays for actomyosin system using a quartz crystal microbalance (QCM). In particular, we report on the difference in the observed resonance frequency and dissipation of a quartz crystal when actin filaments are stationary as opposed to when they are motile. The changes in QCM measurements were studied for various polymer-coated surfaces functionalized with heavy meromyosin (HMM). The results of the QCM experiments show that the HMM-induced sliding velocity of actin filaments is modulated by a combination of the viscoelastic properties of the polymer layer including the HMM motors.
Collapse
Affiliation(s)
- Harm van Zalinge
- Department of Electrical Engineering and Electronics, University of Liverpool, Liverpool, United Kingdom
| | | | | | | | | | | |
Collapse
|
49
|
Heissler SM, Selvadurai J, Bond LM, Fedorov R, Kendrick-Jones J, Buss F, Manstein DJ. Kinetic properties and small-molecule inhibition of human myosin-6. FEBS Lett 2012; 586:3208-14. [PMID: 22884421 PMCID: PMC3527664 DOI: 10.1016/j.febslet.2012.07.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 06/23/2012] [Accepted: 07/10/2012] [Indexed: 11/12/2022]
Abstract
Myosin-6 is an actin-based motor protein that moves its cargo towards the minus-end of actin filaments. Mutations in the gene encoding the myosin-6 heavy chain and changes in the cellular abundance of the protein have been linked to hypertrophic cardiomyopathy, neurodegenerative diseases, and cancer. Here, we present a detailed kinetic characterization of the human myosin-6 motor domain, describe the effect of 2,4,6-triiodophenol on the interaction of myosin-6 with F-actin and nucleotides, and show how addition of the drug reduces the number of myosin-6-dependent vesicle fusion events at the plasma membrane during constitutive secretion.
Collapse
Affiliation(s)
- Sarah M Heissler
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Photoreactive compounds are important tools in life sciences that allow precisely timed covalent crosslinking of ligands and targets. Using a unique technique we have synthesized azidoblebbistatin, which is a derivative of blebbistatin, the most widely used myosin inhibitor. Without UV irradiation azidoblebbistatin exhibits identical inhibitory properties to those of blebbistatin. Using UV irradiation, azidoblebbistatin can be covalently crosslinked to myosin, which greatly enhances its in vitro and in vivo effectiveness. Photo-crosslinking also eliminates limitations associated with the relatively low myosin affinity and water solubility of blebbistatin. The wavelength used for photo-crosslinking is not toxic for cells and tissues, which confers a great advantage in in vivo tests. Because the crosslink results in an irreversible association of the inhibitor to myosin and the irradiation eliminates the residual activity of unbound inhibitor molecules, azidoblebbistatin has a great potential to become a highly effective tool in both structural studies of actomyosin contractility and the investigation of cellular and physiological functions of myosin II. We used azidoblebbistatin to identify previously unknown low-affinity targets of the inhibitor (EC(50) ≥ 50 μM) in Dictyostelium discoideum, while the strongest interactant was found to be myosin II (EC(50) = 5 μM). Our results demonstrate that azidoblebbistatin, and potentially other azidated drugs, can become highly useful tools for the identification of strong- and weak-binding cellular targets and the determination of the apparent binding affinities in in vivo conditions.
Collapse
|