1
|
Wu X, Liu H, Guo YR. Insights into Virus-Host Interactions: Lessons from Caenorhabditis elegans-Orsay Virus Model. Curr Med Sci 2025; 45:169-184. [PMID: 40029496 DOI: 10.1007/s11596-025-00004-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/18/2024] [Accepted: 10/20/2024] [Indexed: 03/05/2025]
Abstract
The study of virus-host interactions has been significantly advanced using model organisms, with nematodes being a prominent example. Caenorhabditis elegans (C. elegans) nematodes have provided valuable insights into the mechanisms of viral infections, host defense strategies, and the development of antiviral therapies. With the discovery of natural viral pathogens of nematodes, Orsay virus, Le Blanc virus, Santeuil virus, and Mělník virus, the exploration of the virus-host interaction model based on nematodes has entered a new era. The virus-host interaction network consists of viruses, hosts, and the antagonistic effects of viruses on host immunity. The nematode virus-host interaction model is a concrete manifestation used to study the complex relationships among these three elements. Previous studies have indicated that during the entire process of nematode infection by viruses, antiviral RNA interference (RNAi) plays a crucial role. Additionally, the host's innate immune responses, such as the antiviral-specific intracellular pathogen response (IPR) and certain signaling pathways homologous to those in humans, are particularly important in the natural immune and antiviral processes of nematodes. These processes are regulated by multiple genes in the host. The reverse genetics system for Orsay virus has been successfully developed to study viral gene function and virus-host interactions. Nematodes serve as simple host models for understanding RNA virus replication, related cellular components, and virus-host interaction mechanisms. These findings will likely contribute to the development of antiviral treatment strategies based on novel targets.
Collapse
Affiliation(s)
- Xun Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Heng Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yusong R Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
2
|
Martina MG, Rubini D, Radi M, Cagno V. Targeting PI4KB and Src/Abl host kinases as broad-spectrum antiviral strategy: Myth or real opportunity? Antiviral Res 2025; 235:106100. [PMID: 39922541 DOI: 10.1016/j.antiviral.2025.106100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Viruses pose a continuous threat to human health. Limited treatment options exist for current viruses, and the risk of infections with newly emerging or re-emerging viruses is increasing. In a pandemic scenario, having a broad-spectrum antiviral to limit viral spread while developing specific antivirals and vaccines is crucial. Targeting host kinases represents a valuable strategy due to the higher barrier to resistance and the broad-spectrum activity it offers. While cells have redundant kinases for the same biological function, viruses rely on specific kinases for their replication cycle, enabling targeted antiviral action with limited toxicity. This review focuses on two extensively studied kinase targets: the lipid kinase phosphatidylinositol 4-kinase IIIβ (PI4KB) and the tyrosine kinase proteins Src and Abl. Compounds active against these targets are reviewed in terms of the viruses they inhibit, their mechanisms of action and their stage of development. While PI4KB inhibitors have reached clinical trials, those targeting Src and Abl remain largely in the preclinical phase. Nevertheless, opportunities exist to improve potency and further understand the specific roles of these kinases in the life cycle of multiple viruses.
Collapse
Affiliation(s)
- Maria Grazia Martina
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy
| | - Daniele Rubini
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy
| | - Marco Radi
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy.
| | - Valeria Cagno
- Institute of Microbiology, University Hospital of Lausanne, University of Lausanne, 1011, Lausanne, Switzerland.
| |
Collapse
|
3
|
Sharafutdinov I, Friedrich B, Rottner K, Backert S, Tegtmeyer N. Cortactin: A major cellular target of viral, protozoal, and fungal pathogens. Mol Microbiol 2024; 122:165-183. [PMID: 38868928 DOI: 10.1111/mmi.15284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/14/2024]
Abstract
Many viral, protozoal, and fungal pathogens represent major human and animal health problems due to their great potential of causing infectious diseases. Research on these pathogens has contributed substantially to our current understanding of both microbial virulence determinants and host key factors during infection. Countless studies have also shed light on the molecular mechanisms of host-pathogen interactions that are employed by these microbes. For example, actin cytoskeletal dynamics play critical roles in effective adhesion, host cell entry, and intracellular movements of intruding pathogens. Cortactin is an eminent host cell protein that stimulates actin polymerization and signal transduction, and recently emerged as fundamental player during host-pathogen crosstalk. Here we review the important role of cortactin as major target for various prominent viral, protozoal and fungal pathogens in humans, and its role in human disease development and cancer progression. Most if not all of these important classes of pathogens have been reported to hijack cortactin during infection through mediating up- or downregulation of cortactin mRNA and protein expression as well as signaling. In particular, pathogen-induced changes in tyrosine and serine phosphorylation status of cortactin at its major phospho-sites (Y-421, Y-470, Y-486, S-113, S-298, S-405, and S-418) are addressed. As has been reported for various Gram-negative and Gram-positive bacteria, many pathogenic viruses, protozoa, and fungi also control these regulatory phospho-sites, for example, by activating kinases such as Src, PAK, ERK1/2, and PKD, which are known to phosphorylate cortactin. In addition, the recruitment of cortactin and its interaction partners, like the Arp2/3 complex and F-actin, to the contact sites between pathogens and host cells is highlighted, as this plays an important role in the infection process and internalization of several pathogens. However, there are also other ways in which the pathogens can exploit the function of cortactin for their needs, as the cortactin-mediated regulation of cellular processes is complex and involves numerous different interaction partners. Here, the current state of knowledge is summarized.
Collapse
Affiliation(s)
- Irshad Sharafutdinov
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Barbara Friedrich
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Klemens Rottner
- Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Steffen Backert
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Nicole Tegtmeyer
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
4
|
Cao L, Huang S, Basant A, Mladenov M, Way M. CK-666 and CK-869 differentially inhibit Arp2/3 iso-complexes. EMBO Rep 2024; 25:3221-3239. [PMID: 39009834 PMCID: PMC11316031 DOI: 10.1038/s44319-024-00201-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/30/2024] [Accepted: 06/18/2024] [Indexed: 07/17/2024] Open
Abstract
The inhibitors, CK-666 and CK-869, are widely used to probe the function of Arp2/3 complex mediated actin nucleation in vitro and in cells. However, in mammals, the Arp2/3 complex consists of 8 iso-complexes, as three of its subunits (Arp3, ArpC1, ArpC5) are encoded by two different genes. Here, we used recombinant Arp2/3 with defined composition to assess the activity of CK-666 and CK-869 against iso-complexes. We demonstrate that both inhibitors prevent linear actin filament formation when ArpC1A- or ArpC1B-containing complexes are activated by SPIN90. In contrast, inhibition of actin branching depends on iso-complex composition. Both drugs prevent actin branch formation by complexes containing ArpC1A, but only CK-869 can inhibit ArpC1B-containing complexes. Consistent with this, in bone marrow-derived macrophages which express low levels of ArpC1A, CK-869 but not CK-666, impacted phagocytosis and cell migration. CK-869 also only inhibits Arp3- but not Arp3B-containing iso-complexes. Our findings have important implications for the interpretation of results using CK-666 and CK-869, given that the relative expression levels of ArpC1 and Arp3 isoforms in cells and tissues remains largely unknown.
Collapse
Affiliation(s)
- LuYan Cao
- The Francis Crick Institute, London, UK.
| | | | | | | | - Michael Way
- The Francis Crick Institute, London, UK.
- Department of Infectious Disease, Imperial College, London, UK.
| |
Collapse
|
5
|
Hernandez-Gonzalez M, Calcraft T, Nans A, Rosenthal PB, Way M. Palisade structure in intact vaccinia virions. mBio 2024; 15:e0313423. [PMID: 38171004 PMCID: PMC10865856 DOI: 10.1128/mbio.03134-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
Vaccinia virus assembly in the cytoplasm of infected cells involves the formation of a biconcave viral core inside the maturing viral particle. The boundary of the core is defined by a pseudohexagonal palisade layer, composed of trimers projecting from an inner wall. To understand the assembly of this complex core architecture, we obtained a subnanometer structure of the palisade trimer by cryo-electron tomography and subtomogram averaging of purified intact virions. Using AlphaFold2 structure predictions, we determined that the palisade is formed from trimers of the proteolytically processed form of the viral protein A10. In addition, we found that each A10 protomer associates with an α-helix (residues 24-66) of A4. Cellular localization assays outside the context of infection demonstrate that the A4 N-terminus is necessary and sufficient to interact with A10. The interaction between A4 and A10 provides insights into how the palisade layer might become tightly associated with the viral membrane during virion maturation. Reconstruction of the palisade layer reveals that, despite local hexagonal ordering, the A10/A4 trimers are widely spaced, suggesting that additional components organize the lattice. This spacing would, however, allow the adoption of the characteristic biconcave shape of the viral core. Finally, we also found that the palisade incorporates multiple copies of a hexameric portal structure. We suggest that these portals are formed by E6, a viral protein that is essential for virion assembly and required to release viral mRNA from the core early in infection.IMPORTANCEPoxviruses such as variola virus (smallpox) and monkeypox cause diseases in humans. Other poxviruses, including vaccinia and modified vaccinia Ankara, are used as vaccine vectors. Given their importance, a greater structural understanding of poxvirus virions is needed. We now performed cryo-electron tomography of purified intact vaccinia virions to study the structure of the palisade, a protein lattice that defines the viral core boundary. We identified the main viral proteins that form the palisade and their interaction surfaces and provided new insights into the organization of the viral core.
Collapse
Affiliation(s)
- Miguel Hernandez-Gonzalez
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Thomas Calcraft
- Structural Biology of Cells and Viruses Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Andrea Nans
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Peter B. Rosenthal
- Structural Biology of Cells and Viruses Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Michael Way
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, United Kingdom
- Department of Infectious Disease, Imperial College, London, United Kingdom
| |
Collapse
|
6
|
Stergas HR, Dillon-Martin M, Dumas CM, Hansen NA, Carasi-Schwartz FJ, D'Amico AR, Finnegan KM, Juch U, Kane KR, Kaplan IE, Masengarb ML, Melero ME, Meyer LE, Sacher CR, Scriven EA, Ebert AM, Ballif BA. CRK and NCK adaptors may functionally overlap in zebrafish neurodevelopment, as indicated by common binding partners and overlapping expression patterns. FEBS Lett 2024; 598:302-320. [PMID: 38058169 DOI: 10.1002/1873-3468.14781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/18/2023] [Accepted: 10/29/2023] [Indexed: 12/08/2023]
Abstract
CRK adaptor proteins are important for signal transduction mechanisms driving cell proliferation and positioning during vertebrate central nervous system development. Zebrafish lacking both CRK family members exhibit small, disorganized retinas with 50% penetrance. The goal of this study was to determine whether another adaptor protein might functionally compensate for the loss of CRK adaptors. Expression patterns in developing zebrafish, and bioinformatic analyses of the motifs recognized by their SH2 and SH3 domains, suggest NCK adaptors are well-positioned to compensate for loss of CRK adaptors. In support of this hypothesis, proteomic analyses found CRK and NCK adaptors share overlapping interacting partners including known regulators of cell adhesion and migration, suggesting their functional intersection in neurodevelopment.
Collapse
Affiliation(s)
| | | | - Caroline M Dumas
- Department of Biology, University of Vermont, Burlington, VT, USA
| | - Nicole A Hansen
- Department of Biology, University of Vermont, Burlington, VT, USA
| | | | - Alex R D'Amico
- Department of Biology, University of Vermont, Burlington, VT, USA
| | - Kylie M Finnegan
- Department of Biology, University of Vermont, Burlington, VT, USA
| | - Uatchet Juch
- Department of Biology, University of Vermont, Burlington, VT, USA
| | - Keeley R Kane
- Department of Biology, University of Vermont, Burlington, VT, USA
| | - Isabel E Kaplan
- Department of Biology, University of Vermont, Burlington, VT, USA
| | | | - Marina E Melero
- Department of Biology, University of Vermont, Burlington, VT, USA
| | - Lauren E Meyer
- Department of Biology, University of Vermont, Burlington, VT, USA
| | - Conrad R Sacher
- Department of Biology, University of Vermont, Burlington, VT, USA
| | - Evan A Scriven
- Department of Biology, University of Vermont, Burlington, VT, USA
| | - Alicia M Ebert
- Department of Biology, University of Vermont, Burlington, VT, USA
| | - Bryan A Ballif
- Department of Biology, University of Vermont, Burlington, VT, USA
| |
Collapse
|
7
|
Lu J, Xing H, Wang C, Tang M, Wu C, Ye F, Yin L, Yang Y, Tan W, Shen L. Mpox (formerly monkeypox): pathogenesis, prevention, and treatment. Signal Transduct Target Ther 2023; 8:458. [PMID: 38148355 PMCID: PMC10751291 DOI: 10.1038/s41392-023-01675-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 12/28/2023] Open
Abstract
In 2022, a global outbreak of Mpox (formerly monkeypox) occurred in various countries across Europe and America and rapidly spread to more than 100 countries and regions. The World Health Organization declared the outbreak to be a public health emergency of international concern due to the rapid spread of the Mpox virus. Consequently, nations intensified their efforts to explore treatment strategies aimed at combating the infection and its dissemination. Nevertheless, the available therapeutic options for Mpox virus infection remain limited. So far, only a few numbers of antiviral compounds have been approved by regulatory authorities. Given the high mutability of the Mpox virus, certain mutant strains have shown resistance to existing pharmaceutical interventions. This highlights the urgent need to develop novel antiviral drugs that can combat both drug resistance and the potential threat of bioterrorism. Currently, there is a lack of comprehensive literature on the pathophysiology and treatment of Mpox. To address this issue, we conducted a review covering the physiological and pathological processes of Mpox infection, summarizing the latest progress of anti-Mpox drugs. Our analysis encompasses approved drugs currently employed in clinical settings, as well as newly identified small-molecule compounds and antibody drugs displaying potential antiviral efficacy against Mpox. Furthermore, we have gained valuable insights from the process of Mpox drug development, including strategies for repurposing drugs, the discovery of drug targets driven by artificial intelligence, and preclinical drug development. The purpose of this review is to provide readers with a comprehensive overview of the current knowledge on Mpox.
Collapse
Affiliation(s)
- Junjie Lu
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Hubei Province, Xiangyang, 441021, China
| | - Hui Xing
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Hubei Province, Xiangyang, 441021, China
| | - Chunhua Wang
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Hubei Province, Xiangyang, 441021, China
| | - Mengjun Tang
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Hubei Province, Xiangyang, 441021, China
| | - Changcheng Wu
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Fan Ye
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Hubei Province, Xiangyang, 441021, China
| | - Lijuan Yin
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Yang Yang
- Shenzhen Key Laboratory of Pathogen and Immunity, National Clinical Research Center for infectious disease, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, 518112, China.
| | - Wenjie Tan
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.
| | - Liang Shen
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Hubei Province, Xiangyang, 441021, China.
| |
Collapse
|
8
|
Basant A, Way M. The amount of Nck rather than N-WASP correlates with the rate of actin-based motility of Vaccinia virus. Microbiol Spectr 2023; 11:e0152923. [PMID: 37855608 PMCID: PMC10883800 DOI: 10.1128/spectrum.01529-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 09/03/2023] [Indexed: 10/20/2023] Open
Abstract
IMPORTANCE Vaccinia virus is a large double-stranded DNA virus and a close relative of Mpox and Variola virus, the causative agent of smallpox. During infection, Vaccinia hijacks its host's transport systems and promotes its spread into neighboring cells by recruiting a signaling network that stimulates actin polymerization. Over the years, Vaccinia has provided a powerful model to understand how signaling networks regulate actin polymerization. Nevertheless, we still lack important quantitative information about the system, including the precise number of viral and host molecules required to induce actin polymerization. Using quantitative fluorescence microscopy techniques, we have determined the number of viral and host signaling proteins accumulating on virions during their egress. Our analysis has uncovered two unexpected new aspects of this process: the number of viral proteins in the virion is not fixed and the velocity of virus movement depends on the level of a single adaptor within the signaling network.
Collapse
Affiliation(s)
- Angika Basant
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute , London, United Kingdom
| | - Michael Way
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute , London, United Kingdom
- Department of Infectious Disease, Imperial College , London, United Kingdom
| |
Collapse
|
9
|
Liu W, Lu JY, Wang YJ, Xu XX, Chen YC, Yu SX, Xiang XW, Chen XZ, Jiu Y, Gao H, Sheng M, Chen ZJ, Hu X, Li D, Maiuri P, Huang X, Ying T, Xu GL, Pang DW, Zhang ZL, Liu B, Liu YJ. Vaccinia virus induces EMT-like transformation and RhoA-mediated mesenchymal migration. J Med Virol 2023; 95:e29041. [PMID: 37621182 DOI: 10.1002/jmv.29041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/17/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023]
Abstract
The emerging outbreak of monkeypox is closely associated with the viral infection and spreading, threatening global public health. Virus-induced cell migration facilitates viral transmission. However, the mechanism underlying this type of cell migration remains unclear. Here we investigate the motility of cells infected by vaccinia virus (VACV), a close relative of monkeypox, through combining multi-omics analyses and high-resolution live-cell imaging. We find that, upon VACV infection, the epithelial cells undergo epithelial-mesenchymal transition-like transformation, during which they lose intercellular junctions and acquire the migratory capacity to promote viral spreading. After transformation, VACV-hijacked RhoA signaling significantly alters cellular morphology and rearranges the actin cytoskeleton involving the depolymerization of robust actin stress fibers, leading-edge protrusion formation, and the rear-edge recontraction, which coordinates VACV-induced cell migration. Our study reveals how poxviruses alter the epithelial phenotype and regulate RhoA signaling to induce fast migration, providing a unique perspective to understand the pathogenesis of poxviruses.
Collapse
Affiliation(s)
- Wei Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Jia-Yin Lu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Ya-Jun Wang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Xin-Xin Xu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Yu-Chen Chen
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Sai-Xi Yu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Xiao-Wei Xiang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Xue-Zhu Chen
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Yaming Jiu
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Hai Gao
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Mengyao Sheng
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Zheng-Jun Chen
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Xinyao Hu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, College of Life Sciences, Institute of Biophysics, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Dong Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, College of Life Sciences, Institute of Biophysics, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Paolo Maiuri
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Xinxin Huang
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Guo-Liang Xu
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, Frontiers Science Center for Cell Responses, College of Chemistry, Nankai University, Tianjin, China
| | - Zhi-Ling Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, China
| | - Baohong Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| | - Yan-Jun Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Bécares M, Albert M, Tárrega C, Coloma R, Falqui M, Luhmann EK, Radoshevich L, Guerra S. ISG15 Is Required for the Dissemination of Vaccinia Virus Extracellular Virions. Microbiol Spectr 2023; 11:e0450822. [PMID: 37036376 PMCID: PMC10269806 DOI: 10.1128/spectrum.04508-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/15/2023] [Indexed: 04/11/2023] Open
Abstract
Viruses have developed many different strategies to counteract immune responses, and Vaccinia virus (VACV) is one of a kind in this aspect. To ensure an efficient infection, VACV undergoes a complex morphogenetic process resulting in the production of two types of infective virions: intracellular mature virus (MV) and extracellular enveloped virus (EV), whose spread depends on different dissemination mechanisms. MVs disseminate after cell lysis, whereas EVs are released or propelled in actin tails from living cells. Here, we show that ISG15 participates in the control of VACV dissemination. Infection of Isg15-/- mouse embryonic fibroblasts with VACV International Health Department-J (IHD-J) strain resulted in decreased EV production, concomitant with reduced induction of actin tails and the abolition of comet-shaped plaque formation, compared to Isg15+/+ cells. Transmission electron microscopy revealed the accumulation of intracellular virus particles and a decrease in extracellular virus particles in the absence of interferon-stimulated gene 15 (ISG15), a finding consistent with altered virus egress. Immunoblot and quantitative proteomic analysis of sucrose gradient-purified virions from both genotypes reported differences in protein levels and composition of viral proteins present on virions, suggesting an ISG15-mediated control of viral proteome. Lastly, the generation of a recombinant IHD-J expressing V5-tagged ISG15 (IHD-J-ISG15) allowed us to identify several viral proteins as potential ISG15 targets, highlighting the proteins A34 and A36, which are essential for EV formation. Altogether, our results indicate that ISG15 is an important host factor in the regulation of VACV dissemination. IMPORTANCE Viral infections are a constant battle between the virus and the host. While the host's only goal is victory, the main purpose of the virus is to spread and conquer new territories at the expense of the host's resources. Along millions of years of incessant encounters, poxviruses have developed a unique strategy consisting in the production two specialized "troops": intracellular mature virions (MVs) and extracellular virions (EVs). MVs mediate transmission between hosts, and EVs ensure advance on the battlefield mediating the long-range dissemination. The mechanism by which the virus "decides" to shed from the primary site of infection and its significant impact in viral transmission is not yet fully established. Here, we demonstrate that this process is finely regulated by ISG15/ISGylation, an interferon-induced ubiquitin-like protein with broad antiviral activity. Studying the mechanism that viruses use during infection could result in new ways of understanding our perpetual war against disease and how we might win the next great battle.
Collapse
Affiliation(s)
- Martina Bécares
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Albert
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Céline Tárrega
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rocío Coloma
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Michela Falqui
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Emma K. Luhmann
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Lilliana Radoshevich
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Susana Guerra
- Department of Preventive Medicine, Public Health and Microbiology, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
11
|
Tang H, Zhang A. Human mpox: Biology, epidemiology, therapeutic options, and development of small molecule inhibitors. Med Res Rev 2023. [PMID: 36891882 DOI: 10.1002/med.21943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 01/22/2023] [Accepted: 02/26/2023] [Indexed: 03/10/2023]
Abstract
Although monkeypox (mpox) has been endemic in Western and Central Africa for 50 years, it has not received sufficient prophylactic and therapeutical attention to avoid evolving into an epidemic. From January 2022 to January 2023, more than 84,000 of mpox cases were reported from 110 countries worldwide. Case numbers appear to be rising every day, making mpox an increasing global public health threat for the foreseeable future. In this perspective, we review the known biology and epidemiology of mpox virus, together with the latest therapeutic options available for mpox treatment. Further, small molecule inhibitors against mpox virus and the future directions in this field are discussed as well.
Collapse
Affiliation(s)
- Hairong Tang
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, and the Engineering Research Center of Cell and Therapeutic Antibody of the Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Ao Zhang
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, and the Engineering Research Center of Cell and Therapeutic Antibody of the Ministry of Education, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China.,Lingang Laboratory, Shanghai, China
| |
Collapse
|
12
|
Hernandez-Gonzalez M, Calcraft T, Nans A, Rosenthal PB, Way M. A succession of two viral lattices drives vaccinia virus assembly. PLoS Biol 2023; 21:e3002005. [PMID: 36862727 PMCID: PMC10013923 DOI: 10.1371/journal.pbio.3002005] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 03/14/2023] [Accepted: 01/19/2023] [Indexed: 03/03/2023] Open
Abstract
During its cytoplasmic replication, vaccinia virus assembles non-infectious spherical immature virions (IV) coated by a viral D13 lattice. Subsequently, IV mature into infectious brick-shaped intracellular mature virions (IMV) that lack D13. Here, we performed cryo-electron tomography (cryo-ET) of frozen-hydrated vaccinia-infected cells to structurally characterise the maturation process in situ. During IMV formation, a new viral core forms inside IV with a wall consisting of trimeric pillars arranged in a new pseudohexagonal lattice. This lattice appears as a palisade in cross-section. As maturation occurs, which involves a 50% reduction in particle volume, the viral membrane becomes corrugated as it adapts to the newly formed viral core in a process that does not appear to require membrane removal. Our study suggests that the length of this core is determined by the D13 lattice and that the consecutive D13 and palisade lattices control virion shape and dimensions during vaccinia assembly and maturation.
Collapse
Affiliation(s)
- Miguel Hernandez-Gonzalez
- Cellular signalling and cytoskeletal function laboratory, The Francis Crick Institute, London, United Kingdom
| | - Thomas Calcraft
- Structural Biology of Cells and Viruses Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Andrea Nans
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Peter B Rosenthal
- Structural Biology of Cells and Viruses Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Michael Way
- Cellular signalling and cytoskeletal function laboratory, The Francis Crick Institute, London, United Kingdom
- Department of Infectious Disease, Imperial College, London, United Kingdom
- * E-mail:
| |
Collapse
|
13
|
Fäßler F, Javoor MG, Datler J, Döring H, Hofer FW, Dimchev G, Hodirnau VV, Faix J, Rottner K, Schur FK. ArpC5 isoforms regulate Arp2/3 complex-dependent protrusion through differential Ena/VASP positioning. SCIENCE ADVANCES 2023; 9:eadd6495. [PMID: 36662867 PMCID: PMC9858492 DOI: 10.1126/sciadv.add6495] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 12/20/2022] [Indexed: 05/10/2023]
Abstract
Regulation of the Arp2/3 complex is required for productive nucleation of branched actin networks. An emerging aspect of regulation is the incorporation of subunit isoforms into the Arp2/3 complex. Specifically, both ArpC5 subunit isoforms, ArpC5 and ArpC5L, have been reported to fine-tune nucleation activity and branch junction stability. We have combined reverse genetics and cellular structural biology to describe how ArpC5 and ArpC5L differentially affect cell migration. Both define the structural stability of ArpC1 in branch junctions and, in turn, by determining protrusion characteristics, affect protein dynamics and actin network ultrastructure. ArpC5 isoforms also affect the positioning of members of the Ena/Vasodilator-stimulated phosphoprotein (VASP) family of actin filament elongators, which mediate ArpC5 isoform-specific effects on the actin assembly level. Our results suggest that ArpC5 and Ena/VASP proteins are part of a signaling pathway enhancing cell migration.
Collapse
Affiliation(s)
- Florian Fäßler
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | | | - Julia Datler
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Hermann Döring
- Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Department of Cell Biology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Florian W. Hofer
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Georgi Dimchev
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | | | - Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Klemens Rottner
- Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Department of Cell Biology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Florian K.M. Schur
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| |
Collapse
|
14
|
Tonnemacher S, Folly-Klan M, Gazi AD, Schäfer S, Pénard E, Eberle R, Kunz R, Walther P, Krijnse Locker J. Vaccinia virus H7-protein is required for the organization of the viral scaffold protein into hexamers. Sci Rep 2022; 12:13007. [PMID: 35906465 PMCID: PMC9338303 DOI: 10.1038/s41598-022-16999-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 07/19/2022] [Indexed: 11/30/2022] Open
Abstract
Viruses of the giant virus family are characterized by a structurally conserved scaffold-capsid protein that shapes the icosahedral virion. The vaccinia virus (VACV) scaffold protein D13, however, transiently shapes the newly assembled viral membrane in to a sphere and is absent from the mature brick-shaped virion. In infected cells D13, a 62 kDa polypeptide, forms trimers that arrange in hexamers and a honey-comb like lattice. Membrane association of the D13-lattice may be mediated by A17, an abundant 21 kDa viral membrane protein. Whether membrane binding mediates the formation of the honey-comb lattice or if other factors are involved, remains elusive. Here we show that H7, a 17 kDa protein conserved among poxviruses, mediates proper formation of D13-hexamers, and hence the honey comb lattice and spherical immature virus. Without H7 synthesis D13 trimers assemble into a large 3D network rather than the typical well organized scaffold layer observed in wild-type infection, composed of short D13 tubes of discrete length that are tightly associated with the endoplasmic reticulum (ER). The data show an unexpected role for H7 in D13 organization and imply that formation of the honey-comb, hexagonal, lattice is essential for VACV membrane assembly and production of infectious progeny. The data are discussed with respect to scaffold proteins of other giant viruses.
Collapse
Affiliation(s)
- Susanne Tonnemacher
- Electron Microscopy of Pathogens, Paul Ehrlich Institute, Paul Ehrlichstreet 51-59, 63225, Langen, Germany
| | - Marcia Folly-Klan
- Ultrastructural Bio-Imaging Unit, Institut Pasteur, 28, rue du Dr. Roux, 75015, Paris, France
| | - Anastasia D Gazi
- Ultrastructural Bio-Imaging Unit, Institut Pasteur, 28, rue du Dr. Roux, 75015, Paris, France
| | - Simon Schäfer
- Electron Microscopy of Pathogens, Paul Ehrlich Institute, Paul Ehrlichstreet 51-59, 63225, Langen, Germany
| | - Esthel Pénard
- Ultrastructural Bio-Imaging Unit, Institut Pasteur, 28, rue du Dr. Roux, 75015, Paris, France
| | - Regina Eberle
- Electron Microscopy of Pathogens, Paul Ehrlich Institute, Paul Ehrlichstreet 51-59, 63225, Langen, Germany
| | - Renate Kunz
- Central Facility for Electron Microscopy, Ulm University, 80981, Ulm, Germany
| | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, 80981, Ulm, Germany
| | - Jacomine Krijnse Locker
- Electron Microscopy of Pathogens, Paul Ehrlich Institute, Paul Ehrlichstreet 51-59, 63225, Langen, Germany. .,Ultrastructural Bio-Imaging Unit, Institut Pasteur, 28, rue du Dr. Roux, 75015, Paris, France. .,Justus Liebig University, Giessen, Germany.
| |
Collapse
|
15
|
Basant A, Way M. The relative binding position of Nck and Grb2 adaptors impacts actin-based motility of Vaccinia virus. eLife 2022; 11:e74655. [PMID: 35796545 PMCID: PMC9333988 DOI: 10.7554/elife.74655] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 07/06/2022] [Indexed: 11/19/2022] Open
Abstract
Phosphotyrosine (pTyr) motifs in unstructured polypeptides orchestrate important cellular processes by engaging SH2-containing adaptors to assemble complex signalling networks. The concept of phase separation has recently changed our appreciation of multivalent networks, however, the role of pTyr motif positioning in their function remains to be explored. We have now investigated this parameter in the operation of the signalling cascade driving actin-based motility and spread of Vaccinia virus. This network involves two pTyr motifs in the viral protein A36 that recruit the adaptors Nck and Grb2 upstream of N-WASP and Arp2/3 complex-mediated actin polymerisation. Manipulating the position of pTyr motifs in A36 and the unrelated p14 from Orthoreovirus, we find that only specific spatial arrangements of Nck and Grb2 binding sites result in robust N-WASP recruitment, Arp2/3 complex driven actin polymerisation and viral spread. This suggests that the relative position of pTyr adaptor binding sites is optimised for signal output. This finding may explain why the relative positions of pTyr motifs are frequently conserved in proteins from widely different species. It also has important implications for regulation of physiological networks, including those undergoing phase transitions.
Collapse
Affiliation(s)
- Angika Basant
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Michael Way
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| |
Collapse
|
16
|
Kramer DA, Piper HK, Chen B. WASP family proteins: Molecular mechanisms and implications in human disease. Eur J Cell Biol 2022; 101:151244. [PMID: 35667337 PMCID: PMC9357188 DOI: 10.1016/j.ejcb.2022.151244] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 02/08/2023] Open
Abstract
Proteins of the Wiskott-Aldrich syndrome protein (WASP) family play a central role in regulating actin cytoskeletal dynamics in a wide range of cellular processes. Genetic mutations or misregulation of these proteins are tightly associated with many diseases. The WASP-family proteins act by transmitting various upstream signals to their conserved WH2-Central-Acidic (WCA) peptide sequence at the C-terminus, which in turn binds to the Arp2/3 complex to stimulate the formation of branched actin networks at membranes. Despite this common feature, the regulatory mechanisms and cellular functions of distinct WASP-family proteins are very different. Here, we summarize and clarify our current understanding of WASP-family proteins and how disruption of their functions is related to human disease.
Collapse
Affiliation(s)
- Daniel A Kramer
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, 2437 Pammel Drive, Ames, IA 50011, USA
| | - Hannah K Piper
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, 2437 Pammel Drive, Ames, IA 50011, USA
| | - Baoyu Chen
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, 2437 Pammel Drive, Ames, IA 50011, USA.
| |
Collapse
|
17
|
Sun J, Zhong X, Fu X, Miller H, Lee P, Yu B, Liu C. The Actin Regulators Involved in the Function and Related Diseases of Lymphocytes. Front Immunol 2022; 13:799309. [PMID: 35371070 PMCID: PMC8965893 DOI: 10.3389/fimmu.2022.799309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/01/2022] [Indexed: 11/21/2022] Open
Abstract
Actin is an important cytoskeletal protein involved in signal transduction, cell structure and motility. Actin regulators include actin-monomer-binding proteins, Wiskott-Aldrich syndrome (WAS) family of proteins, nucleation proteins, actin filament polymerases and severing proteins. This group of proteins regulate the dynamic changes in actin assembly/disassembly, thus playing an important role in cell motility, intracellular transport, cell division and other basic cellular activities. Lymphocytes are important components of the human immune system, consisting of T-lymphocytes (T cells), B-lymphocytes (B cells) and natural killer cells (NK cells). Lymphocytes are indispensable for both innate and adaptive immunity and cannot function normally without various actin regulators. In this review, we first briefly introduce the structure and fundamental functions of a variety of well-known and newly discovered actin regulators, then we highlight the role of actin regulators in T cell, B cell and NK cell, and finally provide a landscape of various diseases associated with them. This review provides new directions in exploring actin regulators and promotes more precise and effective treatments for related diseases.
Collapse
Affiliation(s)
- Jianxuan Sun
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingyu Zhong
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Fu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heather Miller
- Cytek Biosciences, R&D Clinical Reagents, Fremont, CA, United States
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Bing Yu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
18
|
Miyamoto D, Takeuchi K, Chihara K, Fujieda S, Sada K. Protein tyrosine kinase Abl promotes hepatitis C virus particle assembly via interaction with viral substrate activator NS5A. J Biol Chem 2022; 298:101804. [PMID: 35257746 PMCID: PMC8980994 DOI: 10.1016/j.jbc.2022.101804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022] Open
Abstract
Previously, we reported that knockdown of Abl protein tyrosine kinase by shRNA or pharmacological inhibition suppresses particle assembly of J6/JFH1 strain–derived hepatitis C virus (HCV) in Huh-7.5 cells. However, the detailed mechanism by which Abl regulates HCV replication remained unclear. In this study, we established Abl-deficient (Abl−) cells through genome editing and compared HCV production between Abl− cells expressing WT or kinase-dead Abl and parental Huh-7.5 cells. Our findings revealed that Abl expression was not required from the stages of virus attachment and entry to viral gene expression; however, the kinase activity of Abl was necessary for the assembly of HCV particles. Reconstitution experiments using human embryonic kidney 293T cells revealed that phosphorylation of Tyr412 in the activation loop of Abl was enhanced by coexpression with the viral nonstructural protein 5A (NS5A) and was abrogated by the substitution of NS5A Tyr330 with Phe (Y330F), suggesting that NS5A functions as a substrate activator of Abl. Abl–NS5A association was also attenuated by the Y330F mutation of NS5A or the kinase-dead Abl, and Abl Tyr412 phosphorylation was not enhanced by NS5A bearing a mutation disabling homodimerization, although the association of Abl with NS5A was still observed. Taken together, these results demonstrate that Abl forms a phosphorylation-dependent complex with dimeric NS5A necessary for viral particle assembly, but that Abl is capable of complex formation with monomeric NS5A regardless of tyrosine phosphorylation. Our findings provide the foundation of a molecular basis for a new hepatitis C treatment strategy using Abl inhibitors.
Collapse
Affiliation(s)
- Daisuke Miyamoto
- Department of Otorhinolaryngology Head & Neck Surgery, Faculty of Medical Sciences, University of Fukui, Fukui, Japan; Department of Genome Science and Microbiology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Kenji Takeuchi
- Department of Genome Science and Microbiology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan; Organization for Life Science Advancement Programs, University of Fukui, Fukui, Japan
| | - Kazuyasu Chihara
- Department of Genome Science and Microbiology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan; Organization for Life Science Advancement Programs, University of Fukui, Fukui, Japan
| | - Shigeharu Fujieda
- Department of Otorhinolaryngology Head & Neck Surgery, Faculty of Medical Sciences, University of Fukui, Fukui, Japan; Organization for Life Science Advancement Programs, University of Fukui, Fukui, Japan
| | - Kiyonao Sada
- Department of Genome Science and Microbiology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan; Organization for Life Science Advancement Programs, University of Fukui, Fukui, Japan.
| |
Collapse
|
19
|
Agwunobi DO, Wang N, Huang L, Zhang Y, Chang G, Wang K, Li M, Wang H, Liu J. Phosphoproteomic Analysis of Haemaphysalis longicornis Saliva Reveals the Influential Contributions of Phosphoproteins to Blood-Feeding Success. Front Cell Infect Microbiol 2022; 11:769026. [PMID: 35118006 PMCID: PMC8804221 DOI: 10.3389/fcimb.2021.769026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/23/2021] [Indexed: 11/13/2022] Open
Abstract
Tick saliva, an essential chemical secretion of the tick salivary gland, is indispensable for tick survival owing to the physiological influence it exerts on the host defence mechanisms via the instrumentality of its cocktail of pharmacologically active molecules (proteins and peptides). Much research about tick salivary proteome has been performed, but how most of the individual salivary proteins are utilized by ticks to facilitate blood acquisition and pathogen transmission is not yet fully understood. In addition, the phosphorylation of some proteins plays a decisive role in their function. However, due to the low phosphorylation level of protein, especially for a small amount of protein, it is more difficult to study phosphorylation. Maybe, for this reason, the scarcity of works on the phosphorylated tick salivary proteomes still abound. Here, we performed a phosphoproteomic analysis of Haemaphysalis longicornis tick saliva via TiO2 enrichment and the most advanced Thermo Fisher Orbitrap Exploris 480 mass spectrometer for identification. A total of 262 phosphorylated tick saliva proteins were identified and were subjected to functional annotation/enrichment analysis. Cellular and metabolic process terms accounted for the largest proportion of the saliva proteins, with the participation of these proteins in vital intracellular and extracellular transport-oriented processes such as vesicle-mediated transport, exocytic process, cell adhesion, and movement of cell/subcellular component. “Endocytosis”, “Protein processing in endoplasmic reticulum”, and “Purine metabolism” were the most significantly enriched pathways. The knockdown (RNAi) of Tudor domain-containing protein (TCP), actin-depolymerizing factors (ADF), programmed cell death protein (PD), and serine/threonine-protein kinase (SPK) resulted in the dissociation of collagen fibers and the pilosebaceous unit, increased inflammatory infiltrates/granulocytes (possibly heterophiles), and the depletion of the epithelium. Ticks injected with SPK dsRNA engorged normally but with a change in skin colour (possibly an autoimmune reaction) and the failure to produce eggs pointing to a possible role of SPK in reproduction and host immune modulation. Ticks injected with ADF dsRNA failed to acquire blood, underscoring the role of ADF in facilitating tick feeding. The results of this study showed the presence of phosphorylation in tick saliva and highlight the roles of salivary phosphoproteins in facilitating tick feeding.
Collapse
Affiliation(s)
- Desmond O. Agwunobi
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Ningmei Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Lei Huang
- Hebei Xiaowutai Mountain National Nature Reserve Management Center, Zhangjiakou, China
| | - Yefei Zhang
- Hebei Xiaowutai Mountain National Nature Reserve Management Center, Zhangjiakou, China
| | - Guomin Chang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Kuang Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Mengxue Li
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Hui Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
- *Correspondence: Jingze Liu, ; Hui Wang,
| | - Jingze Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, China
- *Correspondence: Jingze Liu, ; Hui Wang,
| |
Collapse
|
20
|
Abstract
The non-catalytic region of tyrosine kinase (Nck) family of adaptors, consisting of Nck1 and Nck2, contributes to selectivity and specificity in the flow of cellular information by recruiting components of signaling networks. Known to play key roles in cytoskeletal remodeling, Nck adaptors modulate host cell-pathogen interactions, immune cell receptor activation, cell adhesion and motility, and intercellular junctions in kidney podocytes. Genetic inactivation of both members of the Nck family results in embryonic lethality; however, viability of mice lacking either one of these adaptors suggests partial functional redundancy. In this Cell Science at a Glance and the accompanying poster, we highlight the molecular organization and functions of the Nck family, focusing on key interactions and pathways, regulation of cellular processes, development, homeostasis and pathogenesis, as well as emerging and non-redundant functions of Nck1 compared to those of Nck2. This article thus aims to provide a timely perspective on the biology of Nck adaptors and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Briana C. Bywaters
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX 7783, USA
| | - Gonzalo M. Rivera
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX 7783, USA
| |
Collapse
|
21
|
Hernandez-Gonzalez M, Larocque G, Way M. Viral use and subversion of membrane organization and trafficking. J Cell Sci 2021; 134:jcs252676. [PMID: 33664154 PMCID: PMC7610647 DOI: 10.1242/jcs.252676] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Membrane trafficking is an essential cellular process conserved across all eukaryotes, which regulates the uptake or release of macromolecules from cells, the composition of cellular membranes and organelle biogenesis. It influences numerous aspects of cellular organisation, dynamics and homeostasis, including nutrition, signalling and cell architecture. Not surprisingly, malfunction of membrane trafficking is linked to many serious genetic, metabolic and neurological disorders. It is also often hijacked during viral infection, enabling viruses to accomplish many of the main stages of their replication cycle, including entry into and egress from cells. The appropriation of membrane trafficking by viruses has been studied since the birth of cell biology and has helped elucidate how this integral cellular process functions. In this Review, we discuss some of the different strategies viruses use to manipulate and take over the membrane compartments of their hosts to promote their replication, assembly and egress.
Collapse
Affiliation(s)
- Miguel Hernandez-Gonzalez
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Gabrielle Larocque
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Michael Way
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Infectious Disease, Imperial College, London W2 1PG, UK
| |
Collapse
|
22
|
Donhauser N, Socher E, Millen S, Heym S, Sticht H, Thoma-Kress AK. Transfer of HTLV-1 p8 and Gag to target T-cells depends on VASP, a novel interaction partner of p8. PLoS Pathog 2020; 16:e1008879. [PMID: 32997728 PMCID: PMC7526893 DOI: 10.1371/journal.ppat.1008879] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 08/12/2020] [Indexed: 12/16/2022] Open
Abstract
The Human T-cell leukemia virus type 1 (HTLV-1) orf I-encoded accessory protein p8 is cleaved from its precursor p12, and both proteins contribute to viral persistence. p8 induces cellular protrusions, which are thought to facilitate transfer of p8 to target cells and virus transmission. Host factors interacting with p8 and mediating p8 transfer are unknown. Here, we report that vasodilator-stimulated phosphoprotein (VASP), which promotes actin filament elongation, is a novel interaction partner of p8 and important for p8 and HTLV-1 Gag cell-to-cell transfer. VASP contains an Ena/VASP homology 1 (EVH1) domain that targets the protein to focal adhesions. Bioinformatics identified a short stretch in p8 (amino acids (aa) 24–45) which may mediate interactions with the EVH1 domain of VASP. Co-immunoprecipitations confirmed interactions of VASP:p8 in 293T, Jurkat and HTLV-1-infected MT-2 cells. Co-precipitation of VASP:p8 could be significantly blocked by peptides mimicking aa 26–37 of p8. Mutational studies revealed that the EVH1-domain of VASP is necessary, but not sufficient for the interaction with p8. Further, deletion of the VASP G- and F-actin binding domains significantly diminished co-precipitation of p8. Imaging identified areas of partial co-localization of VASP with p8 at the plasma membrane and in protrusive structures, which was confirmed by proximity ligation assays. Co-culture experiments revealed that p8 is transferred between Jurkat T-cells via VASP-containing conduits. Imaging and flow cytometry revealed that repression of both endogenous and overexpressed VASP by RNA interference or by CRISPR/Cas9 reduced p8 transfer to the cell surface and to target Jurkat T-cells. Stable repression of VASP by RNA interference in chronically infected MT-2 cells impaired both p8 and HTLV-1 Gag transfer to target Jurkat T-cells, while virus release was unaffected. Thus, we identified VASP as a novel interaction partner of p8, which is important for transfer of HTLV-1 p8 and Gag to target T-cells. The delta-retrovirus Human T-cell leukemia virus type 1 encodes the accessory protein p8, which is generated by proteolytic cleavage from p12. Earlier work has shown that p8 enhances the formation of cellular conduits between T-cells, is transferred through these conduits to target T-cells and increases HTLV-1 transmission. It was suggested that p8 dampens T-cell responses in target T-cells, thus facilitating HTLV-1 infection. Our work sheds light on the mechanism of p8 transfer to target T-cells. We show that vasodilator-stimulated phosphoprotein (VASP), a novel interaction partner of p8, contributes to transfer of p8 to target T-cells. Mechanistically, VASP is crucial for recruitment of p8 to the cell surface. Since VASP is known to promote elongation of actin filaments by preventing them from capping, interactions of p8 with VASP are an elegant strategy to exploit the host cell machinery for being transported to the cell surface, and as a consequence, to other cells. Given that VASP is also important for cell-to-cell transfer of the HTLV-1 Gag protein, our work proposes that VASP is a new cellular target to counteract HTLV-1 cell-to-cell transmission.
Collapse
Affiliation(s)
- Norbert Donhauser
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Eileen Socher
- Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sebastian Millen
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Stefanie Heym
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Heinrich Sticht
- Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Andrea K. Thoma-Kress
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- * E-mail:
| |
Collapse
|
23
|
Abstract
A critical step in the life cycle of a virus is spread to a new target cell, which generally involves the release of new viral particles from the infected cell which can then initiate infection in the next target cell. While cell-free viral particles released into the extracellular environment are necessary for long distance spread, there are disadvantages to this mechanism. These include the presence of immune system components, the low success rate of infection by single particles, and the relative fragility of viral particles in the environment. Several mechanisms of direct cell-to-cell spread have been reported for animal viruses which would avoid the issues associated with cell-free particles. A number of viruses can utilize several different mechanisms of direct cell-to-cell spread, but our understanding of the differential usage by these pathogens is modest. Although the mechanisms of cell-to-cell spread differ among viruses, there is a common exploitation of key pathways and components of the cellular cytoskeleton. Remarkably, some of the viral mechanisms of cell-to-cell spread are surprisingly similar to those used by bacteria. Here we summarize the current knowledge of the conventional and non-conventional mechanisms of viral spread, the common methods used to detect viral spread, and the impact that these mechanisms can have on viral pathogenesis.
Collapse
Affiliation(s)
- Nicolas Cifuentes-Munoz
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, San Miguel, Santiago, Chile
| | - Farah El Najjar
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Rebecca Ellis Dutch
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, United States.
| |
Collapse
|
24
|
Khan AU, Qu R, Fan T, Ouyang J, Dai J. A glance on the role of actin in osteogenic and adipogenic differentiation of mesenchymal stem cells. Stem Cell Res Ther 2020; 11:283. [PMID: 32678016 PMCID: PMC7364498 DOI: 10.1186/s13287-020-01789-2] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/13/2020] [Accepted: 06/23/2020] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have the capacity to differentiate into multiple lineages including osteogenic and adipogenic lineages. An increasing number of studies have indicated that lineage commitment by MSCs is influenced by actin remodeling. Moreover, actin has roles in determining cell shape, nuclear shape, cell spreading, and cell stiffness, which eventually affect cell differentiation. Osteogenic differentiation is promoted in MSCs that exhibit a large spreading area, increased matrix stiffness, higher levels of actin polymerization, and higher density of stress fibers, whereas adipogenic differentiation is prevalent in MSCs with disrupted actin networks. In addition, the mechanical properties of F-actin empower cells to sense and transduce mechanical stimuli, which are also reported to influence differentiation. Various biomaterials, mechanical, and chemical interventions along with pathogen-induced actin alteration in the form of polymerization and depolymerization in MSC differentiation were studied recently. This review will cover the role of actin and its modifications through the use of different methods in inducing osteogenic and adipogenic differentiation.
Collapse
Affiliation(s)
- Asmat Ullah Khan
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Rongmei Qu
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Tingyu Fan
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| |
Collapse
|
25
|
Dimchev G, Amiri B, Humphries AC, Schaks M, Dimchev V, Stradal TEB, Faix J, Krause M, Way M, Falcke M, Rottner K. Lamellipodin tunes cell migration by stabilizing protrusions and promoting adhesion formation. J Cell Sci 2020; 133:jcs239020. [PMID: 32094266 PMCID: PMC7157940 DOI: 10.1242/jcs.239020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 02/19/2020] [Indexed: 01/01/2023] Open
Abstract
Efficient migration on adhesive surfaces involves the protrusion of lamellipodial actin networks and their subsequent stabilization by nascent adhesions. The actin-binding protein lamellipodin (Lpd) is thought to play a critical role in lamellipodium protrusion, by delivering Ena/VASP proteins onto the growing plus ends of actin filaments and by interacting with the WAVE regulatory complex, an activator of the Arp2/3 complex, at the leading edge. Using B16-F1 melanoma cell lines, we demonstrate that genetic ablation of Lpd compromises protrusion efficiency and coincident cell migration without altering essential parameters of lamellipodia, including their maximal rate of forward advancement and actin polymerization. We also confirmed lamellipodia and migration phenotypes with CRISPR/Cas9-mediated Lpd knockout Rat2 fibroblasts, excluding cell type-specific effects. Moreover, computer-aided analysis of cell-edge morphodynamics on B16-F1 cell lamellipodia revealed that loss of Lpd correlates with reduced temporal protrusion maintenance as a prerequisite of nascent adhesion formation. We conclude that Lpd optimizes protrusion and nascent adhesion formation by counteracting frequent, chaotic retraction and membrane ruffling.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Georgi Dimchev
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffen Strasse 7, 38124 Braunschweig, Germany
| | - Behnam Amiri
- Max Delbrück Center for Molecular Medicine, Robert Rössle Strasse 10, 13125 Berlin, Germany
| | - Ashley C Humphries
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Matthias Schaks
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffen Strasse 7, 38124 Braunschweig, Germany
| | - Vanessa Dimchev
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffen Strasse 7, 38124 Braunschweig, Germany
| | - Theresia E B Stradal
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffen Strasse 7, 38124 Braunschweig, Germany
| | - Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Matthias Krause
- Randall Centre of Cell & Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Michael Way
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London NW1 1AT, UK
- Department of Infectious Disease, Imperial College, London W2 1PG, UK
| | - Martin Falcke
- Max Delbrück Center for Molecular Medicine, Robert Rössle Strasse 10, 13125 Berlin, Germany
- Department of Physics, Humboldt University, Newtonstrasse 15, 12489 Berlin, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106 Braunschweig, Germany
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffen Strasse 7, 38124 Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), 38106 Braunschweig, Germany
| |
Collapse
|
26
|
Kühn S, Enninga J. The actin comet guides the way: How
Listeria
actin subversion has impacted cell biology, infection biology and structural biology. Cell Microbiol 2020; 22:e13190. [DOI: 10.1111/cmi.13190] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/04/2020] [Accepted: 02/04/2020] [Indexed: 12/31/2022]
Affiliation(s)
- Sonja Kühn
- Unit of Dynamics of Host‐Pathogen InteractionsInstitut Pasteur Paris France
- Centre National de la Recherche Scientifique (CNRS‐UMR3691) Paris France
| | - Jost Enninga
- Unit of Dynamics of Host‐Pathogen InteractionsInstitut Pasteur Paris France
- Centre National de la Recherche Scientifique (CNRS‐UMR3691) Paris France
| |
Collapse
|
27
|
Abstract
Tunneling nanotubes (TNTs) are actin-based intercellular conduits that connect distant cells and allow intercellular transfer of molecular information, including genetic information, proteins, lipids, and even organelles. Besides providing a means of intercellular communication, TNTs may also be hijacked by pathogens, particularly viruses, to facilitate their spread. Viruses of many different families, including retroviruses, herpesviruses, orthomyxoviruses, and several others have been reported to trigger the formation of TNTs or TNT-like structures in infected cells and use these structures to efficiently spread to uninfected cells. In the current review, we give an overview of the information that is currently available on viruses and TNT-like structures, and we discuss some of the standing questions in this field.
Collapse
|
28
|
Kitamata M, Hotta M, Hamada‐Nakahara S, Suetsugu S. The membrane binding and deformation property of vaccinia virus K1 ankyrin repeat domain protein. Genes Cells 2020; 25:187-196. [DOI: 10.1111/gtc.12749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/11/2020] [Accepted: 01/15/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Manabu Kitamata
- Graduate School of Science and Technology Nara Institute of Science and Technology Ikoma Japan
| | - Mitsukuni Hotta
- Graduate School of Science and Technology Nara Institute of Science and Technology Ikoma Japan
| | | | - Shiro Suetsugu
- Graduate School of Science and Technology Nara Institute of Science and Technology Ikoma Japan
| |
Collapse
|
29
|
Jiang H, Leung C, Tahan S, Wang D. Entry by multiple picornaviruses is dependent on a pathway that includes TNK2, WASL, and NCK1. eLife 2019; 8:50276. [PMID: 31769754 PMCID: PMC6904212 DOI: 10.7554/elife.50276] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 11/25/2019] [Indexed: 12/11/2022] Open
Abstract
Comprehensive knowledge of the host factors required for picornavirus infection would facilitate antiviral development. Here we demonstrate roles for three human genes, TNK2, WASL, and NCK1, in infection by multiple picornaviruses. CRISPR deletion of TNK2, WASL, or NCK1 reduced encephalomyocarditis virus (EMCV), coxsackievirus B3 (CVB3), poliovirus and enterovirus D68 infection, and chemical inhibitors of TNK2 and WASL decreased EMCV infection. Reduced EMCV lethality was observed in mice lacking TNK2. TNK2, WASL, and NCK1 were important in early stages of the viral lifecycle, and genetic epistasis analysis demonstrated that the three genes function in a common pathway. Mechanistically, reduced internalization of EMCV was observed in TNK2 deficient cells demonstrating that TNK2 functions in EMCV entry. Domain analysis of WASL demonstrated that its actin nucleation activity was necessary to facilitate viral infection. Together, these data support a model wherein TNK2, WASL, and NCK1 comprise a pathway important for multiple picornaviruses.
Collapse
Affiliation(s)
- Hongbing Jiang
- Department of Molecular Microbiology, Pathology and Immunology, School of Medicine, Washington University, St. Louis, United States
| | - Christian Leung
- Department of Molecular Microbiology, Pathology and Immunology, School of Medicine, Washington University, St. Louis, United States
| | - Stephen Tahan
- Department of Molecular Microbiology, Pathology and Immunology, School of Medicine, Washington University, St. Louis, United States
| | - David Wang
- Department of Molecular Microbiology, Pathology and Immunology, School of Medicine, Washington University, St. Louis, United States
| |
Collapse
|
30
|
High-Content Analyses of Vaccinia Plaque Formation. Methods Mol Biol 2019. [PMID: 31240682 DOI: 10.1007/978-1-4939-9593-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Vaccinia virus plaque assays are employed for quantification of virus titer through serial dilution of virus on a monolayer of cells. Once the virus titer is diluted enough to allow for only few cells of the monolayer to be infected, clonal spread of infection can be detected by observing the lesion in the cell monolayer or using virus-specific staining methods. Beyond simple titration, plaque formation bares priceless underlying information about subtle virus-host interactions and their impact on virus spread during multiple rounds of infection. These include virus infectivity, mode of virus spread, virus replication rate, and spatiotemporal spread efficacy. How this underlying information can be harnessed using a high-content imaging setup is discussed here.
Collapse
|
31
|
Manhart A, Icheva TA, Guerin C, Klar T, Boujemaa-Paterski R, Thery M, Blanchoin L, Mogilner A. Quantitative regulation of the dynamic steady state of actin networks. eLife 2019; 8:42413. [PMID: 30869077 PMCID: PMC6417862 DOI: 10.7554/elife.42413] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 02/26/2019] [Indexed: 12/30/2022] Open
Abstract
Principles of regulation of actin network dimensions are fundamentally important for cell functions, yet remain unclear. Using both in vitro and in silico approaches, we studied the effect of key parameters, such as actin density, ADF/Cofilin concentration and network width on the network length. In the presence of ADF/Cofilin, networks reached equilibrium and became treadmilling. At the trailing edge, the network disintegrated into large fragments. A mathematical model predicts the network length as a function of width, actin and ADF/Cofilin concentrations. Local depletion of ADF/Cofilin by binding to actin is significant, leading to wider networks growing longer. A single rate of breaking network nodes, proportional to ADF/Cofilin density and inversely proportional to the square of the actin density, can account for the disassembly dynamics. Selective disassembly of heterogeneous networks by ADF/Cofilin controls steering during motility. Our results establish general principles on how the dynamic steady state of actin network emerges from biochemical and structural feedbacks.
Collapse
Affiliation(s)
- Angelika Manhart
- Courant Institute of Mathematical Sciences, New York University, New York, United States.,Department of Biology, New York University, New York, United States
| | - Téa Aleksandra Icheva
- CytomorphoLab, Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, Université Grenoble-Alpes/CEA/CNRS/INRA, Grenoble, France
| | - Christophe Guerin
- CytomorphoLab, Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, Université Grenoble-Alpes/CEA/CNRS/INRA, Grenoble, France
| | - Tobbias Klar
- CytomorphoLab, Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, Université Grenoble-Alpes/CEA/CNRS/INRA, Grenoble, France
| | - Rajaa Boujemaa-Paterski
- CytomorphoLab, Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, Université Grenoble-Alpes/CEA/CNRS/INRA, Grenoble, France
| | - Manuel Thery
- CytomorphoLab, Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, Université Grenoble-Alpes/CEA/CNRS/INRA, Grenoble, France.,CytomorphoLab, Hôpital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, INSERM/AP-HP/Université Paris Diderot, Paris, France
| | - Laurent Blanchoin
- CytomorphoLab, Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, Université Grenoble-Alpes/CEA/CNRS/INRA, Grenoble, France.,CytomorphoLab, Hôpital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, INSERM/AP-HP/Université Paris Diderot, Paris, France
| | - Alex Mogilner
- Courant Institute of Mathematical Sciences, New York University, New York, United States.,Department of Biology, New York University, New York, United States
| |
Collapse
|
32
|
The Ectodomain of the Vaccinia Virus Glycoprotein A34 Is Required for Cell Binding by Extracellular Virions and Contains a Large Region Capable of Interaction with Glycoprotein B5. J Virol 2019; 93:JVI.01343-18. [PMID: 30463966 DOI: 10.1128/jvi.01343-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/12/2018] [Indexed: 12/17/2022] Open
Abstract
An interaction between the orthopoxvirus glycoproteins A34 and B5 has been reported. The transmembrane and ectodomain of A34 are sufficient for interaction with B5, localization of B5 to the site of intracellular wrapping, and subsequent incorporation into the envelope of released extracellular virions. Several mutagenic approaches were undertaken to better define the B5 interaction domain on A34. A set of C-terminal truncations in A34 identified residues 1 to 80 as sufficient for interaction with B5. Additional truncations identified residues 80 to 130 of A34 as sufficient for interaction with B5. To better understand the function of this region, a set of recombinant viruses expressing A34 with the full, partial, or no B5 interaction site (residues 1 to 130, 1 to 100, and 1 to 70, respectively) was constructed. All the recombinants expressing truncations of A34 incorporated B5 into extracellular virions but had a small-plaque phenotype similar to that of a virus with the A34R gene deleted (vΔA34R). Further characterization indicated that the small-plaque phenotype exhibited by these viruses is due to a combination of abrogated actin tail formation, reduced cell binding, and a defect in polyanion-induced nonfusogenic dissolution. Taken together, these results suggest that residues 80 to 130 of A34 are not necessary for the proper localization and incorporation of B5 into extracellular virions and, furthermore, that the C-terminal residues of A34 are involved in cell binding and dissolution.IMPORTANCE Previous studies have shown that the vaccinia virus glycoproteins A34 and B5 interact, and in the absence of A34, B5 is mislocalized and not incorporated into extracellular virions. Here, using a transient-transfection assay, residues 80 to 130 of the ectodomain of A34 were determined to be sufficient for interaction with B5. Recombinant viruses expressing A34 with a full, partial, or no B5 interaction site were constructed and characterized. All of the A34 truncations interacted with B5 as predicted by the transient-transfection studies but had a small-plaque phenotype. Further analysis revealed that all of the recombinants incorporated detectable levels of B5 into released virions but were defective in cell binding and extracellular virion (EV) dissolution. This study is the first to directly demonstrate that A34 is involved in cell binding and implicate the ectodomain in this role.
Collapse
|
33
|
Cell-to-Cell Transmission Is the Main Mechanism Supporting Bovine Viral Diarrhea Virus Spread in Cell Culture. J Virol 2019; 93:JVI.01776-18. [PMID: 30404802 DOI: 10.1128/jvi.01776-18] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 10/24/2018] [Indexed: 12/30/2022] Open
Abstract
After initiation of an infective cycle, spread of virus infection can occur in two fundamentally different ways: (i) viral particles can be released into the external environment and diffuse through the extracellular space until they interact with a new host cell, and (ii) virions can remain associated with infected cells, promoting the direct passage between infected and uninfected cells that is referred to as direct cell-to-cell transmission. Although evidence of cell-associated transmission has accumulated for many different viruses, the ability of members of the genus Pestivirus to use this mode of transmission has not been reported. In the present study, we used a novel recombinant virus expressing the envelope glycoprotein E2 fused to mCherry fluorescent protein to monitor the spreading of bovine viral diarrhea virus (BVDV) (the type member of the pestiviruses) infection. To demonstrate direct cell-to-cell transmission of BVDV, we developed a cell coculture system that allowed us to prove direct transmission from infected to uninfected cells in the presence of neutralizing antibodies. This mode of transmission requires cell-cell contacts and clathrin-mediated receptor-dependent endocytosis. Notably, it overcomes antibody blocking of the BVDV receptor CD46, indicating that cell-to-cell transmission of the virus involves the engagement of coreceptors on the target cell.IMPORTANCE BVDV causes one of the most economically important viral infections for the cattle industry. The virus is able to cross the placenta and infect the fetus, leading to the birth of persistently infected animals, which are reservoirs for the spread of BVDV. The occurrence of persistent infection has hampered the efficacy of vaccination because it requires eliciting levels of protection close to sterilizing immunity to prevent fetal infections. While vaccination prevents disease, BVDV can be detected if animals with neutralizing antibodies are challenged with the virus. Virus cell-to-cell transmission allows the virus to overcome barriers to free virus dissemination, such as antibodies or epithelial barriers. Here we show that BVDV exploits cell-cell contacts to propagate infection in a process that is resistant to antibody neutralization. Our results provide new insights into the mechanisms underlying the pathogenesis of BVDV infection and can aid in the design of effective control strategies.
Collapse
|
34
|
Velle KB, Campellone KG. Enteropathogenic E. coli relies on collaboration between the formin mDia1 and the Arp2/3 complex for actin pedestal biogenesis and maintenance. PLoS Pathog 2018; 14:e1007485. [PMID: 30550556 PMCID: PMC6310289 DOI: 10.1371/journal.ppat.1007485] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/28/2018] [Accepted: 11/23/2018] [Indexed: 12/16/2022] Open
Abstract
Enteropathogenic and enterohemorrhagic E. coli (EPEC and EHEC) are closely related extracellular pathogens that reorganize host cell actin into “pedestals” beneath the tightly adherent bacteria. This pedestal-forming activity is both a critical step in pathogenesis, and it makes EPEC and EHEC useful models for studying the actin rearrangements that underlie membrane protrusions. To generate pedestals, EPEC relies on the tyrosine phosphorylated bacterial effector protein Tir to bind host adaptor proteins that recruit N-WASP, a nucleation-promoting factor that activates the Arp2/3 complex to drive actin polymerization. In contrast, EHEC depends on the effector EspFU to multimerize N-WASP and promote Arp2/3 activation. Although these core pathways of pedestal assembly are well-characterized, the contributions of additional actin nucleation factors are unknown. We investigated potential cooperation between the Arp2/3 complex and other classes of nucleators using chemical inhibitors, siRNAs, and knockout cell lines. We found that inhibition of formins impairs actin pedestal assembly, motility, and cellular colonization for bacteria using the EPEC, but not the EHEC, pathway of actin polymerization. We also identified mDia1 as the formin contributing to EPEC pedestal assembly, as its expression level positively correlates with the efficiency of pedestal formation, and it localizes to the base of pedestals both during their initiation and once they have reached steady state. Collectively, our data suggest that mDia1 enhances EPEC pedestal biogenesis and maintenance by generating seed filaments to be used by the N-WASP-Arp2/3-dependent actin nucleation machinery and by sustaining Src-mediated phosphorylation of Tir. Microbial pathogens that rearrange the host actin cytoskeleton have made valuable contributions to our understanding of cell signaling and movement. The assembly and organization of the actin cytoskeleton is driven by proteins called nucleators, which can be manipulated by bacteria including enteropathogenic Escherichia coli (EPEC), a frequent cause of pediatric diarrhea in developing countries. After ingestion, EPEC adhere tightly to cells of the intestine and hijack the underlying cytoskeleton to create protrusions called actin pedestals. While mechanisms of pedestal assembly involving a nucleator called the Arp2/3 complex have been defined for EPEC, the contribution of additional host nucleators has not been determined. We assessed the roles of several actin nucleators in EPEC pedestals and found that in addition to Arp2/3 complex-mediated nucleation, the formin mDia1 is a key contributor to actin assembly. These findings highlight the importance of nucleator collaboration in pathogenesis, and also advance our understanding of the molecular and cellular basis of EPEC infection, which is ultimately important for the discovery of new drug targets.
Collapse
Affiliation(s)
- Katrina B. Velle
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, United States of America
| | - Kenneth G. Campellone
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
35
|
Higgs HN. A fruitful tree: developing the dendritic nucleation model of actin-based cell motility. Mol Biol Cell 2018. [PMCID: PMC6333179 DOI: 10.1091/mbc.e18-07-0426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
A fundamental question in cell biology concerns how cells move, and this has been the subject of intense research for decades. In the 1990s, a major leap forward was made in our understanding of cell motility, with the proposal of the dendritic nucleation model. This essay describes the events leading to the development of the model, including findings from many laboratories and scientific disciplines. The story is an excellent example of the scientific process in action, with the combination of multiple perspectives leading to robust conclusions.
Collapse
Affiliation(s)
- Henry N. Higgs
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| |
Collapse
|
36
|
Yoon SJ, Park YJ, Kim JS, Lee S, Lee SH, Choi S, Min JK, Choi I, Ryu CM. Pseudomonas syringae evades phagocytosis by animal cells via type III effector-mediated regulation of actin filament plasticity. Environ Microbiol 2018; 20:3980-3991. [PMID: 30251365 DOI: 10.1111/1462-2920.14426] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/18/2018] [Accepted: 09/18/2018] [Indexed: 01/31/2023]
Abstract
Certain animal and plant pathogenic bacteria have developed virulence factors including effector proteins that enable them to overcome host immunity. A plant pathogen, Pseudomonas syringae pv. tomato (Pto) secretes a large repertoire of effectors via a type III secretory apparatus, thereby suppressing plant immunity. Here, we show that Pto causes sepsis in mice. Surprisingly, the effector HopQ1 disrupted animal phagocytosis by inhibiting actin rearrangement via direct interaction with the LIM domain of the animal target protein LIM kinase, a key regulator of actin polymerization. The results provide novel insight into animal host-plant pathogen interactions. In addition, the current study firstly demonstrates that certain plant pathogenic bacteria such as Pto evade phagocytosis by animal cells due to cross-kingdom suppression of host immunity.
Collapse
Affiliation(s)
- Sung-Jin Yoon
- Metabolic Regulation Research Center, KRIBB, Yuseong-gu, Daejeon 34141, South Korea
| | - Young-Jun Park
- Metabolic Regulation Research Center, KRIBB, Yuseong-gu, Daejeon 34141, South Korea.,Department of Functional Genomics, University of Science and Technology (UST), Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jun-Seob Kim
- Infectious Disease Research Center, KRIBB, Yuseong-gu, Daejeon 34141, South Korea
| | - Soohyun Lee
- Infectious Disease Research Center, KRIBB, Yuseong-gu, Daejeon 34141, South Korea
| | - Sang-Hyun Lee
- Biotherapeutics Translational Research Center, KRIBB, Yuseong-gu, Daejeon 34141, South Korea
| | - Song Choi
- Metabolic Regulation Research Center, KRIBB, Yuseong-gu, Daejeon 34141, South Korea
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Center, KRIBB, Yuseong-gu, Daejeon 34141, South Korea
| | - Inpyo Choi
- Department of Functional Genomics, University of Science and Technology (UST), Yuseong-gu, Daejeon, 34141, Republic of Korea.,Immunotherapy Convergence Research Center, KRIBB, Yuseong-gu, Daejeon, 34141, South Korea
| | - Choong-Min Ryu
- Infectious Disease Research Center, KRIBB, Yuseong-gu, Daejeon 34141, South Korea.,Biosystems and Bioengineering Program, University of Science and Technology (UST), Yuseong-gu, Daejeon, 34141, Republic of Korea
| |
Collapse
|
37
|
Pfanzelter J, Mostowy S, Way M. Septins suppress the release of vaccinia virus from infected cells. J Cell Biol 2018; 217:2911-2929. [PMID: 29921601 PMCID: PMC6080921 DOI: 10.1083/jcb.201708091] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 04/08/2018] [Accepted: 05/14/2018] [Indexed: 01/01/2023] Open
Abstract
Septins are conserved components of the cytoskeleton that play important roles in many fundamental cellular processes including division, migration, and membrane trafficking. Septins can also inhibit bacterial infection by forming cage-like structures around pathogens such as Shigella We found that septins are recruited to vaccinia virus immediately after its fusion with the plasma membrane during viral egress. RNA interference-mediated depletion of septins increases virus release and cell-to-cell spread, as well as actin tail formation. Live cell imaging reveals that septins are displaced from the virus when it induces actin polymerization. Septin loss, however, depends on the recruitment of the SH2/SH3 adaptor Nck, but not the activity of the Arp2/3 complex. Moreover, it is the recruitment of dynamin by the third Nck SH3 domain that displaces septins from the virus in a formin-dependent fashion. Our study demonstrates that septins suppress vaccinia release by "entrapping" the virus at the plasma membrane. This antiviral effect is overcome by dynamin together with formin-mediated actin polymerization.
Collapse
Affiliation(s)
- Julia Pfanzelter
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, England, UK
| | - Serge Mostowy
- Section of Microbiology, Medical Research Council Centre for Molecular Bacteriology and Infection, Imperial College London, London, England, UK,Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, England, UK
| | - Michael Way
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, England, UK
| |
Collapse
|
38
|
Stradal TEB, Schelhaas M. Actin dynamics in host-pathogen interaction. FEBS Lett 2018; 592:3658-3669. [PMID: 29935019 PMCID: PMC6282728 DOI: 10.1002/1873-3468.13173] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 06/19/2018] [Accepted: 06/19/2018] [Indexed: 02/06/2023]
Abstract
The actin cytoskeleton and Rho GTPase signaling to actin assembly are prime targets of bacterial and viral pathogens, simply because actin is involved in all motile and membrane remodeling processes, such as phagocytosis, macropinocytosis, endocytosis, exocytosis, vesicular trafficking and membrane fusion events, motility, and last but not least, autophagy. This article aims at providing an overview of the most prominent pathogen‐induced or ‐hijacked actin structures, and an outlook on how future research might uncover additional, equally sophisticated interactions.
Collapse
Affiliation(s)
- Theresia E B Stradal
- Department of Cell Biology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Mario Schelhaas
- Institute of Cellular Virology, ZMBE, University of Münster, Germany
| |
Collapse
|
39
|
Loss of Actin-Based Motility Impairs Ectromelia Virus Release In Vitro but Is Not Critical to Spread In Vivo. Viruses 2018; 10:v10030111. [PMID: 29510577 PMCID: PMC5869504 DOI: 10.3390/v10030111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/01/2018] [Accepted: 03/01/2018] [Indexed: 12/14/2022] Open
Abstract
Ectromelia virus (ECTV) is an orthopoxvirus and the causative agent of mousepox. Like other poxviruses such as variola virus (agent of smallpox), monkeypox virus and vaccinia virus (the live vaccine for smallpox), ECTV promotes actin-nucleation at the surface of infected cells during virus release. Homologs of the viral protein A36 mediate this function through phosphorylation of one or two tyrosine residues that ultimately recruit the cellular Arp2/3 actin-nucleating complex. A36 also functions in the intracellular trafficking of virus mediated by kinesin-1. Here, we describe the generation of a recombinant ECTV that is specifically disrupted in actin-based motility allowing us to examine the role of this transport step in vivo for the first time. We show that actin-based motility has a critical role in promoting the release of virus from infected cells in vitro but plays a minor role in virus spread in vivo. It is likely that loss of microtubule-dependent transport is a major factor for the attenuation observed when A36R is deleted.
Collapse
|
40
|
Huang WYC, Ditlev JA, Chiang HK, Rosen MK, Groves JT. Allosteric Modulation of Grb2 Recruitment to the Intrinsically Disordered Scaffold Protein, LAT, by Remote Site Phosphorylation. J Am Chem Soc 2017; 139:18009-18015. [PMID: 29182244 DOI: 10.1021/jacs.7b09387] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tyrosine phosphorylation of membrane receptors and scaffold proteins followed by recruitment of SH2 domain-containing adaptor proteins constitutes a central mechanism of intracellular signal transduction. During early T-cell receptor (TCR) activation, phosphorylation of linker for activation of T cells (LAT) leading to recruitment of adaptor proteins, including Grb2, is one prototypical example. LAT contains multiple modifiable sites, and this multivalency may provide additional layers of regulation, although this is not well understood. Here, we quantitatively analyze the effects of multivalent phosphorylation of LAT by reconstituting the initial reactions of the TCR signaling pathway on supported membranes. Results from a series of LAT constructs with combinatorial mutations of tyrosine residues reveal a previously unidentified allosteric mechanism in which the binding affinity of LAT:Grb2 depends on the phosphorylation at remote tyrosine sites. Additionally, we find that LAT:Grb2 binding affinity is altered by membrane localization. This allostery mainly regulates the kinetic on-rate, not off-rate, of LAT:Grb2 interactions. LAT is an intrinsically disordered protein, and these data suggest that phosphorylation changes the overall ensemble of configurations to modulate the accessibility of other phosphorylated sites to Grb2. Using Grb2 as a phosphorylation reporter, we further monitored LAT phosphorylation by TCR ζ chain-recruited ZAP-70, which suggests a weakly processive catalysis on membranes. Taken together, these results suggest that signal transmission through LAT is strongly gated and requires multiple phosphorylation events before efficient signal transmission is achieved.
Collapse
Affiliation(s)
- William Y C Huang
- The Howard Hughes Medical Institute Summer Institute, Marine Biological Laboratory , Woods Hole, Massachusetts 02543, United States.,Department of Chemistry, University of California, Berkeley , Berkeley, California 94720, United States
| | - Jonathon A Ditlev
- The Howard Hughes Medical Institute Summer Institute, Marine Biological Laboratory , Woods Hole, Massachusetts 02543, United States.,Department of Biophysics and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center , Dallas, Texas 75390, United States
| | - Han-Kuei Chiang
- Department of Chemistry, University of California, Berkeley , Berkeley, California 94720, United States
| | - Michael K Rosen
- The Howard Hughes Medical Institute Summer Institute, Marine Biological Laboratory , Woods Hole, Massachusetts 02543, United States.,Department of Biophysics and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center , Dallas, Texas 75390, United States
| | - Jay T Groves
- The Howard Hughes Medical Institute Summer Institute, Marine Biological Laboratory , Woods Hole, Massachusetts 02543, United States.,Department of Chemistry, University of California, Berkeley , Berkeley, California 94720, United States
| |
Collapse
|
41
|
Morris DC, Popp JL, Tang LK, Gibbs HC, Schmitt E, Chaki SP, Bywaters BC, Yeh AT, Porter WW, Burghardt RC, Barhoumi R, Rivera GM. Nck deficiency is associated with delayed breast carcinoma progression and reduced metastasis. Mol Biol Cell 2017; 28:3500-3516. [PMID: 28954862 PMCID: PMC5683761 DOI: 10.1091/mbc.e17-02-0106] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 09/15/2017] [Accepted: 09/20/2017] [Indexed: 12/16/2022] Open
Abstract
Nck promotes breast carcinoma progression and metastasis by directing the polarized interaction of carcinoma cells with collagen fibrils, decreasing actin turnover, and enhancing the localization and activity of MMP14 at the cell surface through modulation of the spatiotemporal activation of Cdc42 and RhoA. Although it is known that noncatalytic region of tyrosine kinase (Nck) regulates cell adhesion and migration by bridging tyrosine phosphorylation with cytoskeletal remodeling, the role of Nck in tumorigenesis and metastasis has remained undetermined. Here we report that Nck is required for the growth and vascularization of primary tumors and lung metastases in a breast cancer xenograft model as well as extravasation following injection of carcinoma cells into the tail vein. We provide evidence that Nck directs the polarization of cell–matrix interactions for efficient migration in three-dimensional microenvironments. We show that Nck advances breast carcinoma cell invasion by regulating actin dynamics at invadopodia and enhancing focalized extracellular matrix proteolysis by directing the delivery and accumulation of MMP14 at the cell surface. We find that Nck-dependent cytoskeletal changes are mechanistically linked to enhanced RhoA but restricted spatiotemporal activation of Cdc42. Using a combination of protein silencing and forced expression of wild-type/constitutively active variants, we provide evidence that Nck is an upstream regulator of RhoA-dependent, MMP14-mediated breast carcinoma cell invasion. By identifying Nck as an important driver of breast carcinoma progression and metastasis, these results lay the groundwork for future studies assessing the therapeutic potential of targeting Nck in aggressive cancers.
Collapse
Affiliation(s)
- David C Morris
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| | - Julia L Popp
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| | - Leung K Tang
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| | - Holly C Gibbs
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-4467
| | - Emily Schmitt
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4467
| | - Sankar P Chaki
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| | - Briana C Bywaters
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| | - Alvin T Yeh
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-4467
| | - Weston W Porter
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4467
| | - Robert C Burghardt
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4467
| | - Rola Barhoumi
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4467
| | - Gonzalo M Rivera
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| |
Collapse
|
42
|
Boujemaa-Paterski R, Suarez C, Klar T, Zhu J, Guérin C, Mogilner A, Théry M, Blanchoin L. Network heterogeneity regulates steering in actin-based motility. Nat Commun 2017; 8:655. [PMID: 28935896 PMCID: PMC5608943 DOI: 10.1038/s41467-017-00455-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 06/29/2017] [Indexed: 11/10/2022] Open
Abstract
The growth of branched actin networks powers cell-edge protrusions and motility. A heterogeneous density of actin, which yields to a tunable cellular response, characterizes these dynamic structures. We study how actin organization controls both the rate and the steering during lamellipodium growth. We use a high-resolution surface structuration assay combined with mathematical modeling to describe the growth of a reconstituted lamellipodium. We demonstrate that local monomer depletion at the site of assembly negatively impacts the network growth rate. At the same time, network architecture tunes the protrusion efficiency, and regulates the rate of growth. One consequence of this interdependence between monomer depletion and network architecture effects is the ability of heterogeneous network to impose steering during motility. Therefore, we have established that the general principle, by which the cell can modulate the rate and the direction of a protrusion, is by varying both density and architecture of its actin network. Protrusive cellular structures contain a heterogeneous density of actin, but whether this influences motility is not known. Using an in vitro system and modelling, here the authors show that local actin monomer depletion and network architecture can tune the rate of network growth to impose steering during motility.
Collapse
Affiliation(s)
- Rajaa Boujemaa-Paterski
- CytomorphoLab, Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, Université Grenoble-Alpes/CEA/CNRS/INRA, 38054, Grenoble, France
| | - Cristian Suarez
- CytomorphoLab, Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, Université Grenoble-Alpes/CEA/CNRS/INRA, 38054, Grenoble, France
| | - Tobias Klar
- CytomorphoLab, Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, Université Grenoble-Alpes/CEA/CNRS/INRA, 38054, Grenoble, France
| | - Jie Zhu
- Courant Institute of Mathematical Sciences and Department of Biology, New York University, New York, NY, 10012, USA
| | - Christophe Guérin
- CytomorphoLab, Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, Université Grenoble-Alpes/CEA/CNRS/INRA, 38054, Grenoble, France
| | - Alex Mogilner
- Courant Institute of Mathematical Sciences and Department of Biology, New York University, New York, NY, 10012, USA.
| | - Manuel Théry
- CytomorphoLab, Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, Université Grenoble-Alpes/CEA/CNRS/INRA, 38054, Grenoble, France. .,CytomorphoLab, Hôpital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, INSERM/AP-HP/Université Paris Diderot, 75010, Paris, France.
| | - Laurent Blanchoin
- CytomorphoLab, Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, Université Grenoble-Alpes/CEA/CNRS/INRA, 38054, Grenoble, France. .,CytomorphoLab, Hôpital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, INSERM/AP-HP/Université Paris Diderot, 75010, Paris, France.
| |
Collapse
|
43
|
Durkin CH, Leite F, Cordeiro JV, Handa Y, Arakawa Y, Valderrama F, Way M. RhoD Inhibits RhoC-ROCK-Dependent Cell Contraction via PAK6. Dev Cell 2017; 41:315-329.e7. [PMID: 28486133 PMCID: PMC5425256 DOI: 10.1016/j.devcel.2017.04.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 01/05/2017] [Accepted: 04/12/2017] [Indexed: 01/21/2023]
Abstract
RhoA-mediated regulation of myosin-II activity in the actin cortex controls the ability of cells to contract and bleb during a variety of cellular processes, including cell migration and division. Cell contraction and blebbing also frequently occur as part of the cytopathic effect seen during many different viral infections. We now demonstrate that the vaccinia virus protein F11, which localizes to the plasma membrane, is required for ROCK-mediated cell contraction from 2 hr post infection. Curiously, F11-induced cell contraction is dependent on RhoC and not RhoA signaling to ROCK. Moreover, RhoC-driven cell contraction depends on the upstream inhibition of RhoD signaling by F11. This inhibition prevents RhoD from regulating its downstream effector Pak6, alleviating the suppression of RhoC by the kinase. Our observations with vaccinia have now demonstrated that RhoD recruits Pak6 to the plasma membrane to antagonize RhoC signaling during cell contraction and blebbing. Vaccinia F11 protein is required for virus-induced cell contraction and blebbing F11-induced cell contraction depends on RhoC, but not RhoA, signaling to ROCK RhoD recruits Pak6 to the plasma membrane to antagonize RhoC signaling F11 inhibits RhoD signaling to its downstream effector Pak6
Collapse
Affiliation(s)
- Charlotte H Durkin
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Flavia Leite
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - João V Cordeiro
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Yutaka Handa
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Yoshiki Arakawa
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ferran Valderrama
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Michael Way
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
44
|
Abstract
Many fundamental biological discoveries have been made in Caenorhabditis elegans. The discovery of Orsay virus has enabled studies of host-virus interactions in this model organism. To identify host factors critical for Orsay virus infection, we designed a forward genetic screen that utilizes a virally induced green fluorescent protein (GFP) reporter. Following chemical mutagenesis, two Viro (virus induced reporter off) mutants that failed to express GFP were mapped to sid-3, a nonreceptor tyrosine kinase, and B0280.13 (renamed viro-2), an ortholog of human Wiskott-Aldrich syndrome protein (WASP). Both mutants yielded Orsay virus RNA levels comparable to that of the residual input virus, suggesting that they are not permissive for Orsay virus replication. In addition, we demonstrated that both genes affect an early prereplication stage of Orsay virus infection. Furthermore, it is known that the human ortholog of SID-3, activated CDC42-associated kinase (ACK1/TNK2), is capable of phosphorylating human WASP, suggesting that VIRO-2 may be a substrate for SID-3 in C. elegans. A targeted RNA interference (RNAi) knockdown screen further identified the C. elegans gene nck-1, which has a human ortholog that interacts with TNK2 and WASP, as required for Orsay virus infection. Thus, genetic screening in C. elegans identified critical roles in virus infection for evolutionarily conserved genes in a known human pathway. Orsay virus is the only known virus capable of naturally infecting the model organism Caenorhabditis elegans, which shares many evolutionarily conserved genes with humans. We exploited the robust genetic tractability of C. elegans to identify three host genes, sid-3, viro-2, and nck-1, which are essential for Orsay virus infection. Mutant animals that lack these three genes are highly defective in viral replication. Strikingly, the human orthologs of these three genes, activated CDC42-associated kinase (TNK2), Wiskott-Aldrich syndrome protein (WASP), and noncatalytic region of tyrosine kinase adaptor protein 1 (NCK1) are part of a known signaling pathway in mammals. These results suggest that TNK2, WASP, and NCK1 may play important roles in mammalian virus infection.
Collapse
|
45
|
Zhou T, Li N, Liu S, Jin Y, Fu Q, Gao S, Liu Y, Liu Z. The NCK and ABI adaptor genes in catfish and their involvement in ESC disease response. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 73:119-123. [PMID: 28341353 DOI: 10.1016/j.dci.2017.03.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/20/2017] [Accepted: 03/20/2017] [Indexed: 06/06/2023]
Abstract
Adaptor proteins non-catalytic region of tyrosine kinase (NCK) and Abelson interactor (ABI) are crucial for disease response. NCK1 was identified to be a candidate gene for enteric septicemia of catfish (ESC) disease resistance, and was speculated to play similar roles during ESC and enteropathogenic Escherichia coli (EPEC) pathogenicity. ABI1 was reported as a positional candidate gene for bacterial cold water disease (BCWD) resistance in rainbow trout. In this study, three NCK genes and six ABI genes were identified in the channel catfish (Ictalurus punctatus) genome and blue catfish (I. furcatus) transcriptome, and annotated by domain structures, phylogenetic and syntenic analyses. Their expression patterns were examined in the intestine and liver of catfish after challenge with Edwardsiella ictaluri. In the intestine, NCK1, ABI2a, ABI2b, ABI3a were differentially expressed after E. ictaluri infection. In the liver, NCK2a, NCK2b, ABI1b, ABI2a, ABI2b were significantly upregulated in ESC susceptible fish. In general, the NCK and ABI genes, with exception of ABI3a gene and NCK1 gene, were expressed at higher levels in susceptible fish after infection than in control fish, but were expressed at lower levels in resistant fish than in the control fish. Taken together, these results support the notion that NCK and ABI genes are involved in disease processes facilitating pathogenesis of the E. ictaluri bacteria.
Collapse
Affiliation(s)
- Tao Zhou
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Ning Li
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Shikai Liu
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Yulin Jin
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Qiang Fu
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Sen Gao
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Yang Liu
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Zhanjiang Liu
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
46
|
Carpentier DCJ, Van Loggerenberg A, Dieckmann NMG, Smith GL. Vaccinia virus egress mediated by virus protein A36 is reliant on the F12 protein. J Gen Virol 2017. [PMID: 28631604 PMCID: PMC5656793 DOI: 10.1099/jgv.0.000816] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Egress of vaccinia virus from its host cell is mediated by the microtubule-associated motor kinesin-1, and three viral proteins, A36 and the F12/E2 complex, have been implicated in this process. Deletion of F12 expression causes a more severe reduction in egress than deletion of A36 but whether these proteins are involved in the same or different mechanisms of kinesin-1 recruitment is unknown. Here it is shown that a virus lacking both proteins forms a smaller plaque than mutants lacking either gene alone, indicating non-redundant functions. A36 not only links virions directly to kinesin-1 but also nucleates actin polymerization to propel surface virions away from the host cell. To address the relative importance of these functions for virus spread, a panel of recombinant viruses was constructed in which the ability of A36 to bind kinesin-1 or to nucleate actin polymerization was abrogated individually or together, in the presence or absence of F12 expression. Analysis of these viruses revealed that in the presence of the F12 protein, loss of kinesin-1 interaction made a greater contribution to plaque size than did the formation of actin tails. However in the absence of F12, the ability of A36 to promote egress was abrogated. Therefore, the ability of A36 to promote egress by kinesin-1 is reliant on the F12 protein.
Collapse
Affiliation(s)
- David C J Carpentier
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | | | - Nele M G Dieckmann
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK.,Present address: Cambridge Institute for Medical Research (CIMR), University of Cambridge, Cambridge Biomedical Campus, CB2 0XY, UK
| | - Geoffrey L Smith
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| |
Collapse
|
47
|
Yu-Kemp HC, Kemp JP, Brieher WM. CRMP-1 enhances EVL-mediated actin elongation to build lamellipodia and the actin cortex. J Cell Biol 2017. [PMID: 28630144 PMCID: PMC5551698 DOI: 10.1083/jcb.201606084] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
CRMP proteins regulate the cytoskeleton, but the underlying mechanisms are poorly understood. Yu-Kemp et al. show that CRMP-1 helps Ena/VASP proteins elongate actin filaments to assemble actin networks that are necessary for the integrity of epithelial sheets. Cells can control actin polymerization by nucleating new filaments or elongating existing ones. We recently identified CRMP-1 as a factor that stimulates the formation of Listeria monocytogenes actin comet tails, thereby implicating it in actin assembly. We now show that CRMP-1 is a major contributor to actin assembly in epithelial cells, where it works with the Ena/VASP family member EVL to assemble the actin cytoskeleton in the apical cortex and in protruding lamellipodia. CRMP-1 and EVL bind to one another and together accelerate actin filament barbed-end elongation. CRMP-1 also stimulates actin assembly in the presence of VASP and Mena in vitro, but CRMP-1–dependent actin assembly in MDCK cells is EVL specific. Our results identify CRMP-1 as a novel regulator of actin filament elongation and reveal a surprisingly important role for CRMP-1, EVL, and actin polymerization in maintaining the structural integrity of epithelial sheets.
Collapse
Affiliation(s)
- Hui-Chia Yu-Kemp
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - James P Kemp
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - William M Brieher
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| |
Collapse
|
48
|
Discovery of Novel Small-Molecule Inhibitors of LIM Domain Kinase for Inhibiting HIV-1. J Virol 2017; 91:JVI.02418-16. [PMID: 28381571 PMCID: PMC5469273 DOI: 10.1128/jvi.02418-16] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 03/28/2017] [Indexed: 01/22/2023] Open
Abstract
A dynamic actin cytoskeleton is necessary for viral entry, intracellular migration, and virion release. For HIV-1 infection, during entry, the virus triggers early actin activity by hijacking chemokine coreceptor signaling, which activates a host dependency factor, cofilin, and its kinase, the LIM domain kinase (LIMK). Although knockdown of human LIM domain kinase 1 (LIMK1) with short hairpin RNA (shRNA) inhibits HIV infection, no specific small-molecule inhibitor of LIMK has been available. Here, we describe the design and discovery of novel classes of small-molecule inhibitors of LIMK for inhibiting HIV infection. We identified R10015 as a lead compound that blocks LIMK activity by binding to the ATP-binding pocket. R10015 specifically blocks viral DNA synthesis, nuclear migration, and virion release. In addition, R10015 inhibits multiple viruses, including Zaire ebolavirus (EBOV), Rift Valley fever virus (RVFV), Venezuelan equine encephalitis virus (VEEV), and herpes simplex virus 1 (HSV-1), suggesting that LIMK inhibitors could be developed as a new class of broad-spectrum antiviral drugs. IMPORTANCE The actin cytoskeleton is a structure that gives the cell shape and the ability to migrate. Viruses frequently rely on actin dynamics for entry and intracellular migration. In cells, actin dynamics are regulated by kinases, such as the LIM domain kinase (LIMK), which regulates actin activity through phosphorylation of cofilin, an actin-depolymerizing factor. Recent studies have found that LIMK/cofilin are targeted by viruses such as HIV-1 for propelling viral intracellular migration. Although inhibiting LIMK1 expression blocks HIV-1 infection, no highly specific LIMK inhibitor is available. This study describes the design, medicinal synthesis, and discovery of small-molecule LIMK inhibitors for blocking HIV-1 and several other viruses and emphasizes the feasibility of developing LIMK inhibitors as broad-spectrum antiviral drugs.
Collapse
|
49
|
Flatt JW, Greber UF. Viral mechanisms for docking and delivering at nuclear pore complexes. Semin Cell Dev Biol 2017; 68:59-71. [PMID: 28506891 DOI: 10.1016/j.semcdb.2017.05.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 05/11/2017] [Indexed: 12/22/2022]
Abstract
Some viruses possess the remarkable ability to transport their genomes across nuclear pore complexes (NPCs) for replication inside the host cell's intact nuclear compartment. Viral mechanisms for crossing the restrictive NPC passageway are highly complex and astonishingly diverse, requiring in each case stepwise interaction between incoming virus particles and components of the nuclear transport machinery. Exactly how a large viral genome loaded with accessory proteins is able to pass through the relatively narrow central channel of the NPC without causing catastrophic structural damage is not yet fully understood. It appears likely, however, that the overall structure of the NPC changes in response to the cargo. Translocation may result in nucleic acids being misdelivered to the cytoplasm. Here we consider in detail the diverse strategies that viruses have evolved to target and subvert NPCs during infection. For decades, this process has both captivated and confounded researchers in the fields of virology, cell biology, and structural biology.
Collapse
Affiliation(s)
- Justin W Flatt
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Urs F Greber
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
50
|
Marzook NB, Latham SL, Lynn H, Mckenzie C, Chaponnier C, Grau GE, Newsome TP. Divergent roles of β- and γ-actin isoforms during spread of vaccinia virus. Cytoskeleton (Hoboken) 2017; 74:170-183. [PMID: 28218453 PMCID: PMC7162416 DOI: 10.1002/cm.21356] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 01/31/2017] [Accepted: 02/16/2017] [Indexed: 01/20/2023]
Abstract
Actin is a major component of the cytoskeleton and is present as two isoforms in non‐muscle cells: β‐ and γ‐cytoplasmic actin. These isoforms are strikingly conserved, differing by only four N‐terminal amino acids. During spread from infected cells, vaccinia virus (VACV) particles induce localized actin nucleation that propel virus to surrounding cells and facilitate cell‐to‐cell spread of infection. Here we show that virus‐tipped actin comets are composed of β‐ and γ‐actin. We employed isoform‐specific siRNA knockdown to examine the role of the two isoforms in VACV‐induced actin comets. Despite the high level of similarity between the actin isoforms, and their colocalization, VACV‐induced actin nucleation was dependent exclusively on β‐actin. Knockdown of β‐actin led to a reduction in the release of virus from infected cells, a phenotype dependent on virus‐induced Arp2/3 complex activity. We suggest that local concentrations of actin isoforms may regulate the activity of cellular actin nucleator complexes.
Collapse
Affiliation(s)
- N Bishara Marzook
- School of Life and Environmental Sciences, The University of Sydney, Australia
| | - Sharissa L Latham
- Vascular Immunology Unit, Department of Pathology, School of Medical Sciences & Marie Bashir Institute, The University of Sydney, Australia
| | - Helena Lynn
- School of Life and Environmental Sciences, The University of Sydney, Australia
| | | | - Christine Chaponnier
- Department of Pathology-Immunology, Faculty of Medicine, University of Geneva, Switzerland
| | - Georges E Grau
- Vascular Immunology Unit, Department of Pathology, School of Medical Sciences & Marie Bashir Institute, The University of Sydney, Australia
| | - Timothy P Newsome
- School of Life and Environmental Sciences, The University of Sydney, Australia
| |
Collapse
|