1
|
Amita H, Subair Z, Mora T, Dudhe PE, Dhanasekaran K. Betrayal From the Core: Centriolar and Cytoskeletal Subversion by Infectious Pathogens. Cytoskeleton (Hoboken) 2025. [PMID: 39902598 DOI: 10.1002/cm.22004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/30/2024] [Accepted: 01/24/2025] [Indexed: 02/05/2025]
Abstract
Microbes and parasites have evolved several means to evade and usurp the host cellular machinery to mediate pathogenesis. Being the major microtubule-organizing center (MTOC) of the cell, the centrosome is targeted by multiple viral and nonviral pathogens to mediate their assembly and trafficking within the host cell. This review examines the consequence of such targeting to the centrosome and associated cytoskeletal machinery. We have also amassed a substantial body of evidence of viruses utilizing the cilia within airway epithelium to mediate infection and the hijacking of host cytoskeletal machinery for efficient entry, replication, and egress. While infections have been demonstrated to induce structural, functional, and numerical aberrations in centrosomes, and induce ciliary dysfunction, current literature increasingly supports the notion of a pro-viral role for these organelles. Although less explored, the impact of bacterial and parasitic pathogens on these structures has also been addressed very briefly. Mechanistically, the molecular pathways responsible for these effects remain largely uncharacterized in many instances. Future research focusing on the centriolar triad comprising the centrosome, cilia, and centriolar satellites will undoubtedly provide vital insights into the tactics employed by infectious agents to subvert the host centriole and cytoskeleton-based machinery.
Collapse
Affiliation(s)
- Himanshi Amita
- Laboratory of Centrosome and Cilia Biology, Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Zidhan Subair
- Laboratory of Centrosome and Cilia Biology, Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Tulasiram Mora
- Laboratory of Centrosome and Cilia Biology, Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Pranay Eknath Dudhe
- Laboratory of Centrosome and Cilia Biology, Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Karthigeyan Dhanasekaran
- Laboratory of Centrosome and Cilia Biology, Regional Centre for Biotechnology, Faridabad, Haryana, India
| |
Collapse
|
2
|
Kang G, Roh H. Dual RNA-seq Analyses of Viral Hemorrhagic Septicemia Virus and Olive Flounder (Paralichthys olivaceus) Interactions at Low and High Water Temperatures. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025:105335. [PMID: 39909203 DOI: 10.1016/j.dci.2025.105335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 02/01/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
A vast body of transcriptomic data have considerably contributed to understanding the host responses under viral hemorrhagic septicemia virus (VHSV) infection. However, changes from the perspective of the pathogen and host-pathogen interactions have been relatively underestimated. Given that VHSV genes can be poly-adenylated during replication, this study investigated the global changes from both VHSV and host perspectives using the host transcriptomic data. To achieve this, we utilized VHSV transcriptomic data obtained at different water temperatures (13°C and 20°C). We collected the sequence reads belonging to VHSV through an bioinformatic pipeline developed especially for this study. The VHSV reads were used to construct the consensus reference genome and to investigate the expression of VHSV genes and the frequency of variants under different water temperatures. In addition, both linear and logarithmic scales of VHSV transcription levels, along with host transcriptomes, were used to understand pathogen-host interaction through weighted correlation network analysis (WGCNA). The results revealed that VHSV transcription can exceed 5% of host transcriptome during the infection. Single nucleotide variants (SNVs) appeared more frequently in 13°C groups than in 20°C groups. While VHSV can replicate at both 13°C and 20°C, host transcriptomic responses were notably different, with stronger immune responses and more frequent VHSV genetic changes observed at a lower temperature. This suggests that VHSV infection at low water temperatures significantly influences both the host transcriptional changes and pathogen. Through WGCNA, numerous genes in the module that correlated with VHSV reads on a linear scale were found to be related to cytoskeleton modulation and viral activity. By contrast, the gene module (black) correlated with VHSV reads on a logarithmic scale was strongly associated with host immune responses, such as TNF signaling, necroptosis, and the NF-kappa B signaling pathway. The dual RNA-seq approaches developed in this study will immensely enhance our understanding of host-pathogen interactions across different temperatures.
Collapse
Affiliation(s)
- Gyoungsik Kang
- Department of Aquatic Life Medicine, College of Marine Sciences, Gyeongsang National University, Tongyeong 53064, Republic of Korea
| | - HyeongJin Roh
- Department of Aquatic Life Medicine, College of Marine Sciences, Gyeongsang National University, Tongyeong 53064, Republic of Korea.
| |
Collapse
|
3
|
Lee M, Vetter J, Eichwald C. The influence of the cytoskeleton on the development and behavior of viral factories in mammalian orthoreovirus. Virology 2025; 604:110423. [PMID: 39889480 DOI: 10.1016/j.virol.2025.110423] [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: 09/09/2024] [Revised: 01/06/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Cytosolic viral factories (VFs) of mammalian orthoreovirus (MRV) are sites for viral genome replication and assembly of virus progeny. Despite advancements in reverse genetics, the formation and dynamics of VFs still need to be clarified. MRV exploits host cytoskeletal components like microtubules (MTs) throughout its life cycle, including cell entry, replication, and release. MRV VFs, membrane-less cytosolic inclusions, rely on the viral proteins μ2 and μNS for formation. Protein μ2 interacts and stabilizes MTs through acetylation, supporting VF formation and viral replication, while scaffold protein μNS influences cellular components to aid VF maturation. The disruption of the MT network reduces viral replication, underscoring its importance. Additionally, μ2 associates with MT-organizing centers, modulating the MT dynamics to favor viral replication. In summary, MRV subverts the cytoskeleton to facilitate VF dynamics and promote viral replication and assembly to promote VF dynamics, replication, and assembly, highlighting the critical role of the cytoskeleton in viral replication.
Collapse
Affiliation(s)
- Melissa Lee
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | - Janine Vetter
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
4
|
Hu X, Wang Y, Wang R, Pu Y, Jin R, Nie Y, Shuai X. The hybrid lipoplex induces cytoskeletal rearrangement via autophagy/RhoA signaling pathway for enhanced anticancer gene therapy. Nat Commun 2025; 16:339. [PMID: 39747218 PMCID: PMC11696071 DOI: 10.1038/s41467-024-55727-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 12/21/2024] [Indexed: 01/04/2025] Open
Abstract
Delivering plasmid DNA (pDNA) to solid tumors remains a significant challenge due to the requirement for multiple transport steps and the need to promote delivery efficiency. Herein, we present a virus-mimicking hybrid lipoplex, composed of an arginine-rich cationic lipid, hyaluronic acid derivatives coated gold nanoparticles, and pDNA. This system induces cytoskeletal rearrangements through "outside-in" mechanical and "inside-out" biochemical signaling, overcoming intra- and intercellular barriers to enhance pDNA delivery. By modulating autophagy, RhoA signaling, and cytoskeletal dynamics, we achieve a 20-fold increase in gene expression with high tissue specificity in solid tumors. Furthermore, the system is applied to co-deliver a p53 plasmid and an MDM2 inhibitor, demonstrating significant synergistic antitumor effects in hepatocellular and lung carcinomas.
Collapse
Affiliation(s)
- Xueyi Hu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China
| | - Yichun Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China
| | - Ruohan Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China
| | - Yiyao Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China.
| | - Yu Nie
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, 610064, P. R. China.
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, P. R. China
| |
Collapse
|
5
|
Larrieux A, Sanjuán R. Murine colon cancer derived cells exhibit heterogeneous resistance profiles against an oncolytic virus. Sci Rep 2024; 14:27209. [PMID: 39516525 PMCID: PMC11549347 DOI: 10.1038/s41598-024-78313-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Oncolytic virotherapy has shown efficacy in various animal models and a few human cancers. However, there are still significant limitations for the implementation of these therapies. One such limitation is the emergence of cellular resistances, which may appear rapidly considering the high genetic heterogeneity of most tumors. We previously showed that cellular resistance to an oncolytic virus can be mediated by the chronic activation of innate immunity. Here, we explored the existence of additional resistance mechanisms in murine colon cancer-derived cells. For this purpose, we isolated two cellular clones that were resistant to the oncolytic virus VSV-D51. While one of the clones showed a strong resistance profile associated with increased cytokine-mediated antiviral responses, the other clone showed a lower level of resistance that involves cytoskeletal reorganization, signaling by small GTPases, and cell structural changes. These results demonstrate the capacity of tumor cells to deploy heterogeneous mechanisms of resistance to oncolytic viruses.
Collapse
Affiliation(s)
- Alejandra Larrieux
- Institute for Integrative Systems Biology (I2SysBio), Universitat de València-CSIC, Paterna, 46980, València, Spain
| | - Rafael Sanjuán
- Institute for Integrative Systems Biology (I2SysBio), Universitat de València-CSIC, Paterna, 46980, València, Spain.
| |
Collapse
|
6
|
Chen KR, Yang CY, Shu SG, Lo YC, Lee KW, Wang LC, Chen JB, Shih MC, Chang HC, Hsiao YJ, Wu CL, Tan TH, Ling P. Endosomes serve as signaling platforms for RIG-I ubiquitination and activation. SCIENCE ADVANCES 2024; 10:eadq0660. [PMID: 39504361 PMCID: PMC11540011 DOI: 10.1126/sciadv.adq0660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/01/2024] [Indexed: 11/08/2024]
Abstract
RIG-I-like receptors (RLRs) are cytosolic RNA sensors critical for antiviral immunity. RLR activation is regulated by polyubiquitination and oligomerization following RNA binding. Yet, little is known about how RLRs exploit subcellular organelles to facilitate their posttranslational modifications and activation. Endosomal adaptor TAPE regulates the endosomal TLR and cytosolic RLR pathways. The potential interplay between RIG-I signaling and endosomes has been explored. Here, we report that endosomes act as platforms for facilitating RIG-I polyubiquitination and complex formation. RIG-I was translocated onto endosomes to form signaling complexes upon activation. Ablation of endosomes impaired RIG-I signaling to type I IFN activation. TAPE mediates the interaction and polyubiquitination of RIG-I and TRIM25. TAPE-deficient myeloid cells were defective in type I IFN activation upon RNA ligand and virus challenges. Myeloid TAPE deficiency increased the susceptibility to RNA virus infection in vivo. Our work reveals endosomes as signaling platforms for RIG-I activation and antiviral immunity.
Collapse
Affiliation(s)
- Kuan-Ru Chen
- Department of Microbiology and Immunology, National Cheng Kung University, 70101 Tainan, Taiwan
- Department of Medical Research, E-Da Hospital, I-Shou University, 824005 Kaohsiung, Taiwan
| | - Chia-Yu Yang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, 33302 Tao-Yuan, Taiwan
| | - San-Ging Shu
- Department of Pediatrics, Ditmanson Medical Foundation Chia-Yi Christian Hospital, 60002 Chiayi City, Taiwan
| | - Yin-Chiu Lo
- Department of Microbiology and Immunology, National Cheng Kung University, 70101 Tainan, Taiwan
| | - Kuan-Wei Lee
- Department of Microbiology and Immunology, National Cheng Kung University, 70101 Tainan, Taiwan
| | - Li-Chun Wang
- Department of Microbiology and Immunology, National Cheng Kung University, 70101 Tainan, Taiwan
| | - Jia-Bao Chen
- Department of Microbiology and Immunology, National Cheng Kung University, 70101 Tainan, Taiwan
| | - Meng-Cen Shih
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, 70101 Tainan, Taiwan
| | - Hung-Chun Chang
- Department of Microbiology and Immunology, National Cheng Kung University, 70101 Tainan, Taiwan
| | - Yu-Ju Hsiao
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, 70101 Tainan, Taiwan
| | - Chao-Liang Wu
- Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, 60002 Chiayi City, Taiwan
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Pin Ling
- Department of Microbiology and Immunology, National Cheng Kung University, 70101 Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, 70101 Tainan, Taiwan
| |
Collapse
|
7
|
Hofmann N, Bartkuhn M, Becker S, Biedenkopf N, Böttcher-Friebertshäuser E, Brinkrolf K, Dietzel E, Fehling SK, Goesmann A, Heindl MR, Hoffmann S, Karl N, Maisner A, Mostafa A, Kornecki L, Müller-Kräuter H, Müller-Ruttloff C, Nist A, Pleschka S, Sauerhering L, Stiewe T, Strecker T, Wilhelm J, Wuerth JD, Ziebuhr J, Weber F, Schmitz ML. Distinct negative-sense RNA viruses induce a common set of transcripts encoding proteins forming an extensive network. J Virol 2024; 98:e0093524. [PMID: 39283124 PMCID: PMC11494938 DOI: 10.1128/jvi.00935-24] [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: 05/29/2024] [Accepted: 08/14/2024] [Indexed: 10/23/2024] Open
Abstract
The large group of negative-strand RNA viruses (NSVs) comprises many important pathogens. To identify conserved patterns in host responses, we systematically compared changes in the cellular RNA levels after infection of human hepatoma cells with nine different NSVs of different virulence degrees. RNA sequencing experiments indicated that the amount of viral RNA in host cells correlates with the number of differentially expressed host cell transcripts. Time-resolved differential gene expression analysis revealed a common set of 178 RNAs that are regulated by all NSVs analyzed. A newly developed open access web application allows downloads and visualizations of all gene expression comparisons for individual viruses over time or between several viruses. Most of the genes included in the core set of commonly differentially expressed genes (DEGs) encode proteins that serve as membrane receptors, signaling proteins and regulators of transcription. They mainly function in signal transduction and control immunity, metabolism, and cell survival. One hundred sixty-five of the DEGs encode host proteins from which 47 have already been linked to the regulation of viral infections in previous studies and 89 proteins form a complex interaction network that may function as a core hub to control NSV infections.IMPORTANCEThe infection of cells with negative-strand RNA viruses leads to the differential expression of many host cell RNAs. The differential spectrum of virus-regulated RNAs reflects a large variety of events including anti-viral responses, cell remodeling, and cell damage. Here, these virus-specific differences and similarities in the regulated RNAs were measured in a highly standardized model. A newly developed app allows interested scientists a wide range of comparisons and visualizations.
Collapse
Affiliation(s)
- Nina Hofmann
- Bioinformatics and Systems Biology, Justus Liebig University Giessen, Giessen, Germany
| | - Marek Bartkuhn
- Biomedical Informatics and Systems Medicine Science Unit for Basic and Clinical Medicine, Justus Liebig University Giessen, Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University Giessen, Giessen, Germany
| | - Stephan Becker
- Institute of Virology, Philipps-University Marburg, Hans-Meerwein-Straße, Marburg, Germany
| | - Nadine Biedenkopf
- Institute of Virology, Philipps-University Marburg, Hans-Meerwein-Straße, Marburg, Germany
| | | | - Karina Brinkrolf
- Bioinformatics and Systems Biology, Justus Liebig University Giessen, Giessen, Germany
| | - Erik Dietzel
- Institute of Virology, Philipps-University Marburg, Hans-Meerwein-Straße, Marburg, Germany
| | | | - Alexander Goesmann
- Bioinformatics and Systems Biology, Justus Liebig University Giessen, Giessen, Germany
| | - Miriam Ruth Heindl
- Institute of Virology, Philipps-University Marburg, Hans-Meerwein-Straße, Marburg, Germany
| | - Simone Hoffmann
- Institute for Virology, FB10-Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Nadja Karl
- Institute of Medical Virology, FB11-Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Andrea Maisner
- Institute of Virology, Philipps-University Marburg, Hans-Meerwein-Straße, Marburg, Germany
| | - Ahmed Mostafa
- Institute of Medical Virology, FB11-Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Laura Kornecki
- Institute of Virology, Philipps-University Marburg, Hans-Meerwein-Straße, Marburg, Germany
| | - Helena Müller-Kräuter
- Institute of Virology, Philipps-University Marburg, Hans-Meerwein-Straße, Marburg, Germany
| | - Christin Müller-Ruttloff
- Institute of Medical Virology, FB11-Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Andrea Nist
- Genomics Core Facility, Institute of Molecular Oncology, Philipps University of Marburg, Marburg, Germany
| | - Stephan Pleschka
- Institute of Medical Virology, FB11-Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Lucie Sauerhering
- Institute of Virology, Philipps-University Marburg, Hans-Meerwein-Straße, Marburg, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Philipps University of Marburg, Marburg, Germany
| | - Thomas Strecker
- Institute of Virology, Philipps-University Marburg, Hans-Meerwein-Straße, Marburg, Germany
| | - Jochen Wilhelm
- Institute for Lung Health (ILH), Justus Liebig University Giessen, Giessen, Germany
| | - Jennifer D. Wuerth
- Institute for Virology, FB10-Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - John Ziebuhr
- Institute of Medical Virology, FB11-Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Friedemann Weber
- Institute for Virology, FB10-Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - M. Lienhard Schmitz
- Institute of Biochemistry, FB11-Medicine, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
8
|
Aïqui-Reboul-Paviet O, Bakhache W, Bernard E, Holsteyn L, Neyret A, Briant L. The Rac1-PAK1-Arp2/3 signaling axis regulates CHIKV nsP1-induced filopodia and optimal viral genome replication. J Virol 2024; 98:e0061224. [PMID: 39297643 PMCID: PMC11495065 DOI: 10.1128/jvi.00612-24] [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: 04/05/2024] [Accepted: 07/10/2024] [Indexed: 10/23/2024] Open
Abstract
Alphavirus infection induces dramatic remodeling of host cellular membranes, producing filopodia-like and intercellular extensions. The formation of filopodia-like extensions has been primarily assigned to the replication protein nsP1, which binds and reshapes the host plasma membrane when expressed alone. While reported decades ago, the molecular mechanisms behind nsP1 membrane deformation remain unknown. Using mammalian epithelial cells and Chikungunya virus (CHIKV) as models, we characterized nsP1-induced membrane deformations as highly dynamic actin-rich lamellipodia and filopodia-like extensions. Through pharmacological inhibition and genetic invalidation, we identified the critical contribution of the Rac1 GTPase and its downstream effectors PAK1 and the actin nucleator Arp2 in nsP1-induced membrane deformation. An intact Rac1-PAK1-Arp2 signaling axis was also required for optimal CHIKV genome replication. Therefore, our results designate the Rac1-PAK1-Arp2 pathway as an essential signaling node for CHIKV infection and establish a parallel requirement for host factors involved in nsP1-induced plasma membrane reshaping and assembly of a functional replication complex.IMPORTANCEThe alphavirus nsP1 protein dramatically remodels host cellular membranes, resulting in the formation of filopodia-like extensions. Although described decades ago, the molecular mechanisms controlling these membrane deformations and their functional importance remain elusive. Our study provides mechanistic insight, uncovering the critical role of the Rac1 GTPase, along with its downstream effectors PAK1 and the actin nucleator Arp2, in the nsP1-associated phenotype. Furthermore, we demonstrate that the Rac1-PAK1-Arp2 pathway is essential for optimal CHIKV genome replication. Our findings establish a parallel in the cellular mechanisms governing nsP1-induced plasma membrane reshaping and the production of a functional replication complex in infected cells.
Collapse
Affiliation(s)
| | - William Bakhache
- RNA Viruses and Metabolism Team, IRIM-CNRS UMR9004, Montpellier, France
| | - Eric Bernard
- RNA Viruses and Metabolism Team, IRIM-CNRS UMR9004, Montpellier, France
| | - Lise Holsteyn
- RNA Viruses and Metabolism Team, IRIM-CNRS UMR9004, Montpellier, France
| | - Aymeric Neyret
- RNA Viruses and Metabolism Team, IRIM-CNRS UMR9004, Montpellier, France
| | - Laurence Briant
- RNA Viruses and Metabolism Team, IRIM-CNRS UMR9004, Montpellier, France
| |
Collapse
|
9
|
Bessenay A, Bisio H, Belmudes L, Couté Y, Bertaux L, Claverie JM, Abergel C, Jeudy S, Legendre M. Complex transcriptional regulations of a hyperparasitic quadripartite system in giant viruses infecting protists. Nat Commun 2024; 15:8608. [PMID: 39384766 PMCID: PMC11464507 DOI: 10.1038/s41467-024-52906-1] [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: 05/23/2024] [Accepted: 09/25/2024] [Indexed: 10/11/2024] Open
Abstract
Hyperparasitism is a common pattern in nature that is not limited to cellular organisms. Giant viruses infecting protists can be hyperparasitized by smaller ones named virophages. In addition, both may carry episomal DNA molecules known as transpovirons in their particles. They all share transcriptional regulatory elements that dictate the expression of their genes within viral factories built by giant viruses in the host cytoplasm. This suggests the existence of interactions between their respective transcriptional networks. Here we investigated Acanthamoeba castellanii cells infected by a giant virus (megavirus chilensis), and coinfected with a virophage (zamilon vitis) and/or a transpoviron (megavirus vitis transpoviron). Infectious cycles were monitored through time-course RNA sequencing to decipher the transcriptional program of each partner and its impact on the gene expression of the others. We found highly diverse transcriptional responses. While the giant virus drastically reshaped the host cell transcriptome, the transpoviron had no effect on the gene expression of any of the players. In contrast, the virophage strongly modified the giant virus gene expression, albeit transiently, without altering the protein composition of mature viral particles. The virophage also induced the overexpression of transpoviron genes, likely through the indirect upregulation of giant virus-encoded transcription factors. Together, these analyses document the intricated transcriptionally regulated networks taking place in the infected cell.
Collapse
Affiliation(s)
- Alexandra Bessenay
- Aix-Marseille University, Centre National de la Recherche Scientifique, Information Génomique & Structurale (IGS), Unité Mixte de Recherche 7256 (Institut de Microbiologie de la Méditerranée, FR3479), IM2B, IOM, Marseille, Cedex 9, France
| | - Hugo Bisio
- Aix-Marseille University, Centre National de la Recherche Scientifique, Information Génomique & Structurale (IGS), Unité Mixte de Recherche 7256 (Institut de Microbiologie de la Méditerranée, FR3479), IM2B, IOM, Marseille, Cedex 9, France
| | - Lucid Belmudes
- Univ. Grenoble Alpes, INSERM, CEA, UA13 BGE, CNRS, CEA, FR2048, Grenoble, France
| | - Yohann Couté
- Univ. Grenoble Alpes, INSERM, CEA, UA13 BGE, CNRS, CEA, FR2048, Grenoble, France
| | - Lionel Bertaux
- Aix-Marseille University, Centre National de la Recherche Scientifique, Information Génomique & Structurale (IGS), Unité Mixte de Recherche 7256 (Institut de Microbiologie de la Méditerranée, FR3479), IM2B, IOM, Marseille, Cedex 9, France
- Aix-Marseille University, Centre National de la Recherche Scientifique, Laboratoire de Chimie Bactérienne (LCB), Unité Mixte de Recherche 7283 (Institut de Microbiologie de la Méditerranée, FR3479), IM2B, Marseille, France
| | - Jean-Michel Claverie
- Aix-Marseille University, Centre National de la Recherche Scientifique, Information Génomique & Structurale (IGS), Unité Mixte de Recherche 7256 (Institut de Microbiologie de la Méditerranée, FR3479), IM2B, IOM, Marseille, Cedex 9, France
| | - Chantal Abergel
- Aix-Marseille University, Centre National de la Recherche Scientifique, Information Génomique & Structurale (IGS), Unité Mixte de Recherche 7256 (Institut de Microbiologie de la Méditerranée, FR3479), IM2B, IOM, Marseille, Cedex 9, France
| | - Sandra Jeudy
- Aix-Marseille University, Centre National de la Recherche Scientifique, Information Génomique & Structurale (IGS), Unité Mixte de Recherche 7256 (Institut de Microbiologie de la Méditerranée, FR3479), IM2B, IOM, Marseille, Cedex 9, France.
| | - Matthieu Legendre
- Aix-Marseille University, Centre National de la Recherche Scientifique, Information Génomique & Structurale (IGS), Unité Mixte de Recherche 7256 (Institut de Microbiologie de la Méditerranée, FR3479), IM2B, IOM, Marseille, Cedex 9, France.
| |
Collapse
|
10
|
Martin MF, Bonaventure B, McCray NE, Peersen OB, Rozen-Gagnon K, Stapleford KA. Distinct chikungunya virus polymerase palm subdomains contribute to viral protein accumulation and virion production. PLoS Pathog 2024; 20:e1011972. [PMID: 39401243 PMCID: PMC11501042 DOI: 10.1371/journal.ppat.1011972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 10/24/2024] [Accepted: 09/22/2024] [Indexed: 10/26/2024] Open
Abstract
Alphaviruses encode an error-prone RNA-dependent RNA polymerase (RdRp), nsP4, required for genome synthesis, yet how the RdRp functions in the complete alphavirus life cycle is not well-defined. Previous work using chikungunya virus has established the importance of the nsP4 residue cysteine 483 in replication. Given the location of residue C483 in the nsP4 palm domain, we hypothesized that other residues within this domain and surrounding subdomains would also contribute to polymerase function. To test this hypothesis, we designed a panel of nsP4 variants via homology modeling based on the coxsackievirus B3 3D polymerase. We rescued each variant in mammalian and mosquito cells and discovered that the palm domain and ring finger subdomain contribute to host-specific replication. In C6/36 cells, we found that while the nsP4 variants had replicase function similar to that of wild-type CHIKV, many variants presented changes in protein accumulation and virion production even when viral nonstructural and structural proteins were produced. Finally, we found that WT CHIKV and nsP4 variant replication and protein production could be enhanced in mammalian cells at 28°C, yet growing virus under these conditions led to changes in virus infectivity. Taken together, these studies highlight that distinct nsP4 subdomains are required for proper RNA transcription and translation, having major effects on virion production.
Collapse
Affiliation(s)
- Marie-France Martin
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Boris Bonaventure
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Nia E. McCray
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Olve B. Peersen
- Department of Biochemistry & Molecular Biology, Colorado State University, Fort Collins, Colorado, United States of America
| | | | - Kenneth A. Stapleford
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, United States of America
| |
Collapse
|
11
|
Aoki-Utsubo C, Kameoka M, Deng L, Hanafi M, Dewi BE, Sudarmono P, Wakita T, Hotta H. Statins enhance extracellular release of hepatitis C virus particles through ERK5 activation. Microbiol Immunol 2024; 68:359-370. [PMID: 39073705 DOI: 10.1111/1348-0421.13166] [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: 04/29/2024] [Revised: 06/13/2024] [Accepted: 07/09/2024] [Indexed: 07/30/2024]
Abstract
Statins, such as lovastatin, have been known to inhibit 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase. Statins were reported to moderately suppress hepatitis C virus (HCV) replication in cultured cells harboring HCV RNA replicons. We report here using an HCV cell culture (HCVcc) system that high concentrations of lovastatin (5-20 μg/mL) markedly enhanced the release of HCV infectious particles (virion) in the culture supernatants by up to 40 times, without enhancing HCV RNA replication, HCV protein synthesis, or HCV virion assembly in the cells. We also found that lovastatin increased the phosphorylation (activation) level of extracellular-signal-regulated kinase 5 (ERK5) in both the infected and uninfected cells in a dose-dependent manner. The lovastatin-mediated increase of HCV virion release was partially reversed by selective ERK5 inhibitors, BIX02189 and XMD8-92, or by ERK5 knockdown using small interfering RNA (siRNA). Moreover, we demonstrated that other cholesterol-lowering statins, but not dehydrolovastatin that is incapable of inhibiting HMG-CoA reductase and activating ERK5, enhanced HCV virion release to the same extent as observed with lovastatin. These results collectively suggest that statins markedly enhance HCV virion release from infected cells through HMG-CoA reductase inhibition and ERK5 activation.
Collapse
Affiliation(s)
- Chie Aoki-Utsubo
- Department of Public Health, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Masanori Kameoka
- Department of Public Health, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Lin Deng
- Division of Infectious Disease Control, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Muhammad Hanafi
- Research Center for Chemistry, National Research and Innovation Agency (BRIN), Serpong, Indonesia
| | - Beti Ernawati Dewi
- Department of Microbiology, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Pratiwi Sudarmono
- Department of Microbiology, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Takaji Wakita
- National Institute of Infectious Diseases, Tokyo, Japan
| | - Hak Hotta
- Department of Public Health, Graduate School of Health Sciences, Kobe University, Kobe, Japan
- Faculty of Clinical Nutrition and Dietetics, Konan Women's University, Kobe, Japan
| |
Collapse
|
12
|
Bowers DF, Ciano KA. F-actin and heparan sulfate proteoglycan distribution in female mosquito salivary glands and ducts. JOURNAL OF MEDICAL ENTOMOLOGY 2024; 61:1214-1221. [PMID: 39001615 DOI: 10.1093/jme/tjae060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/18/2023] [Accepted: 07/09/2024] [Indexed: 09/12/2024]
Abstract
Directly involved in the "suck-and-spit" physiology, female mosquito salivary glands (SGs) primarily imbibe blood for egg development and release anticoagulants to keep blood flowing. Indirectly involved, mosquitoes can uptake arboviruses during blood feeding from a viremic host. This research examined the presence of the filamentous cytoplasmic contractile protein (F-actin) and heparan sulfate proteoglycan (HSPG), in the female mosquito SGs. Immunofluorescent antibody labeling of actin molecules or HSPG combined with anatomy suggests that F-actin forms a network in the SG lobe parenchymal cells attached to intralobar ducts by HSPG. In addition, F-actin twists around intralobar SG ducts in a beaded manner, altogether involved in the expulsion of SG secretions. This arrangement in female Aedes aegypti SGs, suggests that F-actin structures are integrally involved in transmitting infectious agents into hosts.
Collapse
|
13
|
Raab JE, Hamilton DJ, Harju TB, Huynh TN, Russo BC. Pushing boundaries: mechanisms enabling bacterial pathogens to spread between cells. Infect Immun 2024; 92:e0052423. [PMID: 38661369 PMCID: PMC11385730 DOI: 10.1128/iai.00524-23] [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] [Indexed: 04/26/2024] Open
Abstract
For multiple intracellular bacterial pathogens, the ability to spread directly into adjacent epithelial cells is an essential step for disease in humans. For pathogens such as Shigella, Listeria, Rickettsia, and Burkholderia, this intercellular movement frequently requires the pathogens to manipulate the host actin cytoskeleton and deform the plasma membrane into structures known as protrusions, which extend into neighboring cells. The protrusion is then typically resolved into a double-membrane vacuole (DMV) from which the pathogen quickly escapes into the cytosol, where additional rounds of intercellular spread occur. Significant progress over the last few years has begun to define the mechanisms by which intracellular bacterial pathogens spread. This review highlights the interactions of bacterial and host factors that drive mechanisms required for intercellular spread with a focus on how protrusion structures form and resolve.
Collapse
Affiliation(s)
- Julie E. Raab
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Desmond J. Hamilton
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Tucker B. Harju
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Thao N. Huynh
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Brian C. Russo
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| |
Collapse
|
14
|
Pérez-Moro C, Sáez C, Sifres A, López C, Dhillon NPS, Picó B, Pérez-de-Castro A. Genetic Dissection of ToLCNDV Resistance in Resistant Sources of Cucumis melo. Int J Mol Sci 2024; 25:8880. [PMID: 39201567 PMCID: PMC11354858 DOI: 10.3390/ijms25168880] [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: 06/27/2024] [Revised: 08/07/2024] [Accepted: 08/12/2024] [Indexed: 09/02/2024] Open
Abstract
Tomato leaf curl New Delhi virus (ToLCNDV) is a begomovirus causing significant melon (Cucumis melo) crop losses globally. This study aims to map the ToLCNDV resistance in the PI 414723 melon accession, previously identified and characterized through phenotypic studies, thereby exploring shared genomic regions with the established resistant source WM-7. In the present study, WM-7 and PI 414723 were crossed with the susceptible accessions 'Rochet' and 'Blanco' respectively, to generate F1 hybrids. These hybrids were self-pollinated to generate the populations for mapping the ToLCNDV resistance region and designing markers for marker-assisted selection. Disease evaluation included visual symptom scoring, viral-load quantification and tissue printing. Genotyping-by-sequencing and SNP markers were used for quantitative trait loci (QTL) mapping. For genetic analysis, qPCR and bulked segregant RNA-seq (BSR-seq) were performed. Gene expression was assessed using RNA-seq, and qRT-PCR was used for confirmation. The research narrows the candidate region for resistance in WM-7 and identifies overlapping QTLs on chromosome 11 in PI 414723, found in the region of the DNA primase large subunit. BSR-seq and expression analyses highlight potential regulatory roles of chromosome 2 in conferring resistance. Differential expression was confirmed for six genes in the candidate region on chromosome 2. This study confirms the existence of common resistance genes in PI 414723 and WM-7.
Collapse
Affiliation(s)
- Clara Pérez-Moro
- Instituto de Conservación y Mejora de la Agrodiversidad Valenciana (COMAV-UPV), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain; (C.P.-M.); (C.S.); (A.S.); (C.L.)
| | - Cristina Sáez
- Instituto de Conservación y Mejora de la Agrodiversidad Valenciana (COMAV-UPV), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain; (C.P.-M.); (C.S.); (A.S.); (C.L.)
- Centro de Biotecnología y Genómica de Plantas UPM-INIA and E.T.S. Ingeniería Agronómica, Alimentaria y de Biosistemas, Universidad Politécnica de Madrid, 28031 Madrid, Spain
| | - Alicia Sifres
- Instituto de Conservación y Mejora de la Agrodiversidad Valenciana (COMAV-UPV), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain; (C.P.-M.); (C.S.); (A.S.); (C.L.)
| | - Carmelo López
- Instituto de Conservación y Mejora de la Agrodiversidad Valenciana (COMAV-UPV), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain; (C.P.-M.); (C.S.); (A.S.); (C.L.)
| | - Narinder P. S. Dhillon
- World Vegetable Center, East and Southeast Asia, Kasetsart University, Kamphaeng Saen, Nakhon Pathom 73140, Thailand;
| | - Belén Picó
- Instituto de Conservación y Mejora de la Agrodiversidad Valenciana (COMAV-UPV), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain; (C.P.-M.); (C.S.); (A.S.); (C.L.)
| | - Ana Pérez-de-Castro
- Instituto de Conservación y Mejora de la Agrodiversidad Valenciana (COMAV-UPV), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain; (C.P.-M.); (C.S.); (A.S.); (C.L.)
| |
Collapse
|
15
|
Fujimoto A, Kawai H, Kawamura R, Kitamura A. Interaction of Receptor-Binding Domain of the SARS-CoV-2 Omicron Variant with hACE2 and Actin. Cells 2024; 13:1318. [PMID: 39195208 PMCID: PMC11352305 DOI: 10.3390/cells13161318] [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: 07/03/2024] [Revised: 08/01/2024] [Accepted: 08/04/2024] [Indexed: 08/29/2024] Open
Abstract
The omicron variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was identified in 2021 as a variant with heavy amino acid mutations in the spike protein, which is targeted by most vaccines, compared to previous variants. Amino acid substitutions in the spike proteins may alter their affinity for host viral receptors and the host interactome. Here, we found that the receptor-binding domain (RBD) of the omicron variant of SARS-CoV-2 exhibited an increased affinity for human angiotensin-converting enzyme 2, a viral cell receptor, compared to the prototype RBD. Moreover, we identified β- and γ-actin as omicron-specific binding partners of RBD. Protein complex predictions revealed that many omicron-specific amino acid substitutions affected the affinity between RBD of the omicron variant and actin. Our findings indicate that proteins localized to different cellular compartments exhibit strong binding to the omicron RBD.
Collapse
Affiliation(s)
- Ai Fujimoto
- Laboratory of Cellular and Molecular Sciences, Graduate School of Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Hokkaido, Japan; (A.F.)
| | - Haruki Kawai
- Laboratory of Cellular and Molecular Sciences, Graduate School of Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Hokkaido, Japan; (A.F.)
| | - Rintaro Kawamura
- Laboratory of Cellular and Molecular Sciences, Graduate School of Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Hokkaido, Japan; (A.F.)
| | - Akira Kitamura
- Laboratory of Cellular and Molecular Sciences, Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Hokkaido, Japan
- PRIME, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
16
|
Hotter D, Kunzelmann M, Kiefer F, Leukhardt C, Fackler C, Jäger S, Solzin J. High-Throughput Determination of Infectious Virus Titers by Kinetic Measurement of Infection-Induced Changes in Cell Morphology. Int J Mol Sci 2024; 25:8076. [PMID: 39125646 PMCID: PMC11311753 DOI: 10.3390/ijms25158076] [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: 06/03/2024] [Revised: 07/18/2024] [Accepted: 07/21/2024] [Indexed: 08/12/2024] Open
Abstract
Infectivity assays are the key analytical technology for the development and manufacturing of virus-based therapeutics. Here, we introduce a novel assay format that utilizes label-free bright-field images to determine the kinetics of infection-dependent changes in cell morphology. In particular, cell rounding is directly proportional to the amount of infectious virus applied, enabling rapid determination of viral titers in relation to a standard curve. Our kinetic infectious virus titer (KIT) assay is stability-indicating and, due to its sensitive readout method, provides results within 24 h post-infection. Compared to traditional infectivity assays, which depend on a single readout of an infection endpoint, cumulated analysis of kinetic data by a fit model results in precise results (CV < 20%) based on only three wells per sample. This approach allows for a high throughput with ~400 samples processed by a single operator per week. We demonstrate the applicability of the KIT assay for the genetically engineered oncolytic VSV-GP, Newcastle disease virus (NDV), and parapoxvirus ovis (ORFV), but it can potentially be extended to a wide range of viruses that induce morphological changes upon infection. The versatility of this assay, combined with its independence from specific instruments or software, makes it a promising solution to overcome the analytical bottleneck in infectivity assays within the pharmaceutical industry and as a routine method in academic research.
Collapse
Affiliation(s)
- Dominik Hotter
- Boehringer Ingelheim Pharma GmbH & Co. KG, Viral Therapeutics Center, 88397 Biberach an der Riss, Germany
| | - Marco Kunzelmann
- Boehringer Ingelheim Pharma GmbH & Co. KG, Development Biologicals, 88397 Biberach an der Riss, Germany
| | - Franziska Kiefer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Viral Therapeutics Center, 88397 Biberach an der Riss, Germany
| | - Chiara Leukhardt
- Boehringer Ingelheim Pharma GmbH & Co. KG, Viral Therapeutics Center, 88397 Biberach an der Riss, Germany
| | - Carolin Fackler
- Boehringer Ingelheim Pharma GmbH & Co. KG, Viral Therapeutics Center, 88397 Biberach an der Riss, Germany
| | - Stefan Jäger
- Boehringer Ingelheim Pharma GmbH & Co. KG, Central Nervous System Diseases Research, 88397 Biberach an der Riss, Germany
| | - Johannes Solzin
- Boehringer Ingelheim Pharma GmbH & Co. KG, Viral Therapeutics Center, 88397 Biberach an der Riss, Germany
| |
Collapse
|
17
|
Lin Y, Exell J, Lin H, Zhang C, Welsher KD. Hour-Long, Kilohertz Sampling Rate Three-Dimensional Single-Virus Tracking in Live Cells Enabled by StayGold Fluorescent Protein Fusions. J Phys Chem B 2024; 128:5590-5600. [PMID: 38808440 DOI: 10.1021/acs.jpcb.4c01710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
A viral infection process covers a large range of spatiotemporal scales. Tracking the viral infection process with fluorescent labels over long durations while maintaining a fast sampling rate requires bright and highly photostable labels. StayGold is a recently identified green fluorescent protein that has a greater photostability and higher signal intensity under identical illumination conditions compared to existing fluorescence protein variants. Here, StayGold protein fusions were used to generate virus-like particles (StayGold-VLPs) to achieve hour-long 3D single-virus tracking (SVT) with 1000 localizations per second (kHz sampling rate) in live cells. The expanded photon budget from StayGold protein fusions prolonged the tracking duration, facilitating a comprehensive study of viral trafficking dynamics with high temporal resolution over long time scales. The development of StayGold-VLPs presents a simple and general VLP labeling strategy for better performance in SVT, enabling exponentially more information to be collected from single trajectories and allowing for the future possibility of observing the entire life cycle of a single virus.
Collapse
Affiliation(s)
- Yuxin Lin
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Jack Exell
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Haoting Lin
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Chen Zhang
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Kevin D Welsher
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| |
Collapse
|
18
|
Lin Y, Exell J, Lin H, Zhang C, Welsher KD. Hour-long, Kilohertz Sampling Rate 3D Single-virus Tracking in Live Cells Enabled by StayGold Fluorescent Protein Fusions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.14.585070. [PMID: 38559049 PMCID: PMC10979972 DOI: 10.1101/2024.03.14.585070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The viral infection process covers a large range of spatiotemporal scales. Tracking the viral infection process with fluorescent labels over long durations while maintaining a fast sampling rate requires bright and highly photostable labels. StayGold is a recently identified green fluorescent protein that has a greater photostability and higher signal intensity under identical illumination conditions as compared to existing fluorescence protein variants. Here, StayGold protein fusions were used to generate virus-like particles (StayGold-VLPs) to achieve hour-long 3D single-virus tracking (SVT) with one thousand localizations per second (kHz sampling rate) in live cells. The expanded photon budget from StayGold protein fusions prolonged the tracking duration, facilitating a comprehensive study of viral trafficking dynamics with high temporal resolution over long timescales. The development of StayGold-VLPs presents a simple and general VLP labeling strategy for better performance in SVT, enabling exponentially more information to be collected from single trajectories and allowing for the future possibility of observing the whole life cycle of a single virus.
Collapse
Affiliation(s)
- Yuxin Lin
- Department of Chemistry, Duke University; Durham, North Carolina 27708, United States
| | - Jack Exell
- Department of Chemistry, Duke University; Durham, North Carolina 27708, United States
| | - Haoting Lin
- Department of Chemistry, Duke University; Durham, North Carolina 27708, United States
| | - Chen Zhang
- Department of Chemistry, Duke University; Durham, North Carolina 27708, United States
| | - Kevin D. Welsher
- Department of Chemistry, Duke University; Durham, North Carolina 27708, United States
| |
Collapse
|
19
|
Erban T, Kadleckova D, Sopko B, Harant K, Talacko P, Markovic M, Salakova M, Kadlikova K, Tachezy R, Tachezy J. Varroa destructor parasitism and Deformed wing virus infection in honey bees are linked to peroxisome-induced pathways. Proteomics 2024; 24:e2300312. [PMID: 38446070 DOI: 10.1002/pmic.202300312] [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: 08/17/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/07/2024]
Abstract
The ectoparasitic mite Varroa destructor transmits and triggers viral infections that have deleterious effects on honey bee colonies worldwide. We performed a manipulative experiment in which worker bees collected at emergence were exposed to Varroa for 72 h, and their proteomes were compared with those of untreated control bees. Label-free quantitative proteomics identified 77 differentially expressed A. mellifera proteins (DEPs). In addition, viral proteins were identified by orthogonal analysis, and most importantly, Deformed wing virus (DWV) was found at high levels/intensity in Varroa-exposed bees. Pathway enrichment analysis suggested that the main pathways affected included peroxisomal metabolism, cyto-/exoskeleton reorganization, and cuticular proteins. Detailed examination of individual DEPs revealed that additional changes in DEPs were associated with peroxisomal function. In addition, the proteome data support the importance of TGF-β signaling in Varroa-DWV interaction and the involvement of the mTORC1 and Hippo pathways. These results suggest that the effect of DWV on bees associated with Varroa feeding results in aberrant autophagy. In particular, autophagy is selectively modulated by peroxisomes, to which the observed proteome changes strongly corresponded. This study complements previous research with different study designs and suggests the importance of the peroxisome, which plays a key role in viral infections.
Collapse
Affiliation(s)
- Tomas Erban
- Proteomics and Metabolomics Laboratory, Crop Research Institute, Prague 6-Ruzyne, Czechia
| | - Dominika Kadleckova
- Department of Genetics and Microbiology, Faculty of Science BIOCEV, Charles University, Vestec, Czechia
| | - Bruno Sopko
- Proteomics and Metabolomics Laboratory, Crop Research Institute, Prague 6-Ruzyne, Czechia
| | - Karel Harant
- Proteomics Core Facility, Faculty of Science BIOCEV, Charles University, Vestec, Czechia
| | - Pavel Talacko
- Proteomics Core Facility, Faculty of Science BIOCEV, Charles University, Vestec, Czechia
| | - Martin Markovic
- Proteomics and Metabolomics Laboratory, Crop Research Institute, Prague 6-Ruzyne, Czechia
| | - Martina Salakova
- Department of Genetics and Microbiology, Faculty of Science BIOCEV, Charles University, Vestec, Czechia
| | - Klara Kadlikova
- Proteomics and Metabolomics Laboratory, Crop Research Institute, Prague 6-Ruzyne, Czechia
| | - Ruth Tachezy
- Department of Genetics and Microbiology, Faculty of Science BIOCEV, Charles University, Vestec, Czechia
| | - Jan Tachezy
- Department of Parasitology, Faculty of Science BIOCEV, Charles University, Vestec, Czechia
| |
Collapse
|
20
|
Stephens C, Naghavi MH. The host cytoskeleton: a key regulator of early HIV-1 infection. FEBS J 2024; 291:1835-1848. [PMID: 36527282 PMCID: PMC10272291 DOI: 10.1111/febs.16706] [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: 10/21/2022] [Revised: 11/29/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
Due to its central role in cell biology, the cytoskeleton is a key regulator of viral infection, influencing nearly every step of the viral life cycle. In this review, we will discuss the role of two key components of the cytoskeleton, namely the actin and microtubule networks in early HIV-1 infection. We will discuss key contributions to processes ranging from the attachment and entry of viral particles at the cell surface to their arrival and import into the nucleus and identify areas where further research into this complex relationship may yield new insights into HIV-1 pathogenesis.
Collapse
Affiliation(s)
- Christopher Stephens
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Mojgan H. Naghavi
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| |
Collapse
|
21
|
Vetter J, Lee M, Eichwald C. The Role of the Host Cytoskeleton in the Formation and Dynamics of Rotavirus Viroplasms. Viruses 2024; 16:668. [PMID: 38793550 PMCID: PMC11125917 DOI: 10.3390/v16050668] [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: 03/24/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Rotavirus (RV) replicates within viroplasms, membraneless electron-dense globular cytosolic inclusions with liquid-liquid phase properties. In these structures occur the virus transcription, replication, and packaging of the virus genome in newly assembled double-layered particles. The viroplasms are composed of virus proteins (NSP2, NSP5, NSP4, VP1, VP2, VP3, and VP6), single- and double-stranded virus RNAs, and host components such as microtubules, perilipin-1, and chaperonins. The formation, coalescence, maintenance, and perinuclear localization of viroplasms rely on their association with the cytoskeleton. A stabilized microtubule network involving microtubules and kinesin Eg5 and dynein molecular motors is associated with NSP5, NSP2, and VP2, facilitating dynamic processes such as viroplasm coalescence and perinuclear localization. Key post-translation modifications, particularly phosphorylation events of RV proteins NSP5 and NSP2, play pivotal roles in orchestrating these interactions. Actin filaments also contribute, triggering the formation of the viroplasms through the association of soluble cytosolic VP4 with actin and the molecular motor myosin. This review explores the evolving understanding of RV replication, emphasizing the host requirements essential for viroplasm formation and highlighting their dynamic interplay within the host cell.
Collapse
Affiliation(s)
| | | | - Catherine Eichwald
- Institute of Virology, University of Zurich, 8057 Zurich, Switzerland; (J.V.); (M.L.)
| |
Collapse
|
22
|
Shi Q, Zhao R, Chen L, Liu T, Di T, Zhang C, Zhang Z, Wang F, Han Z, Sun J, Liu S. Newcastle disease virus activates diverse signaling pathways via Src to facilitate virus entry into host macrophages. J Virol 2024; 98:e0191523. [PMID: 38334327 PMCID: PMC10949470 DOI: 10.1128/jvi.01915-23] [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: 12/08/2023] [Accepted: 12/27/2023] [Indexed: 02/10/2024] Open
Abstract
As an intrinsic cellular mechanism responsible for the internalization of extracellular ligands and membrane components, caveolae-mediated endocytosis (CavME) is also exploited by certain pathogens for endocytic entry [e.g., Newcastle disease virus (NDV) of paramyxovirus]. However, the molecular mechanisms of NDV-induced CavME remain poorly understood. Herein, we demonstrate that sialic acid-containing gangliosides, rather than glycoproteins, were utilized by NDV as receptors to initiate the endocytic entry of NDV into HD11 cells. The binding of NDV to gangliosides induced the activation of a non-receptor tyrosine kinase, Src, leading to the phosphorylation of caveolin-1 (Cav1) and dynamin-2 (Dyn2), which contributed to the endocytic entry of NDV. Moreover, an inoculation of cells with NDV-induced actin cytoskeletal rearrangement through Src to facilitate NDV entry via endocytosis and direct fusion with the plasma membrane. Subsequently, unique members of the Rho GTPases family, RhoA and Cdc42, were activated by NDV in a Src-dependent manner. Further analyses revealed that RhoA and Cdc42 regulated the activities of specific effectors, cofilin and myosin regulatory light chain 2, responsible for actin cytoskeleton rearrangement, through diverse intracellular signaling cascades. Taken together, our results suggest that an inoculation of NDV-induced Src-mediated cellular activation by binding to ganglioside receptors. This process orchestrated NDV endocytic entry by modulating the activities of caveolae-associated Cav1 and Dyn2, as well as specific Rho GTPases and downstream effectors. IMPORTANCE In general, it is known that the paramyxovirus gains access to host cells through direct penetration at the plasma membrane; however, emerging evidence suggests more complex entry mechanisms for paramyxoviruses. The endocytic entry of Newcastle disease virus (NDV), a representative member of the paramyxovirus family, into multiple types of cells has been recently reported. Herein, we demonstrate the binding of NDV to induce ganglioside-activated Src signaling, which is responsible for the endocytic entry of NDV through caveolae-mediated endocytosis. This process involved Src-dependent activation of the caveolae-associated Cav1 and Dyn2, as well as specific Rho GTPase and downstream effectors, thereby orchestrating the endocytic entry process of NDV. Our findings uncover a novel molecular mechanism of endocytic entry of NDV into host cells and provide novel insight into paramyxovirus mechanisms of entry.
Collapse
Affiliation(s)
- Qiankai Shi
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Ran Zhao
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Linna Chen
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Tianyi Liu
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Tao Di
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Chunwei Zhang
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhiying Zhang
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Fangfang Wang
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zongxi Han
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Junfeng Sun
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Shengwang Liu
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
23
|
Pushalkar S, Wu S, Maity S, Pressler M, Rendleman J, Vitrinel B, Jeffery L, Abdelhadi R, Chen M, Ross T, Carlock M, Choi H, Vogel C. Complex changes in serum protein levels in COVID-19 convalescents. Sci Rep 2024; 14:4479. [PMID: 38396092 PMCID: PMC10891133 DOI: 10.1038/s41598-024-54534-7] [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: 11/03/2023] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
The COVID-19 pandemic, triggered by severe acute respiratory syndrome coronavirus 2, has affected millions of people worldwide. Much research has been dedicated to our understanding of COVID-19 disease heterogeneity and severity, but less is known about recovery associated changes. To address this gap in knowledge, we quantified the proteome from serum samples from 29 COVID-19 convalescents and 29 age-, race-, and sex-matched healthy controls. Samples were acquired within the first months of the pandemic. Many proteins from pathways known to change during acute COVID-19 illness, such as from the complement cascade, coagulation system, inflammation and adaptive immune system, had returned to levels seen in healthy controls. In comparison, we identified 22 and 15 proteins with significantly elevated and lowered levels, respectively, amongst COVID-19 convalescents compared to healthy controls. Some of the changes were similar to those observed for the acute phase of the disease, i.e. elevated levels of proteins from hemolysis, the adaptive immune systems, and inflammation. In contrast, some alterations opposed those in the acute phase, e.g. elevated levels of CETP and APOA1 which function in lipid/cholesterol metabolism, and decreased levels of proteins from the complement cascade (e.g. C1R, C1S, and VWF), the coagulation system (e.g. THBS1 and VWF), and the regulation of the actin cytoskeleton (e.g. PFN1 and CFL1) amongst COVID-19 convalescents. We speculate that some of these shifts might originate from a transient decrease in platelet counts upon recovery from the disease. Finally, we observed race-specific changes, e.g. with respect to immunoglobulins and proteins related to cholesterol metabolism.
Collapse
Affiliation(s)
- Smruti Pushalkar
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA.
| | - Shaohuan Wu
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Shuvadeep Maity
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
- Birla Institute of Technology and Science-Pilani (BITS Pilani), Hyderabad, India
| | - Matthew Pressler
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Justin Rendleman
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Burcu Vitrinel
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Lauren Jeffery
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Ryah Abdelhadi
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Mechi Chen
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Ted Ross
- Cleveland Clinic Florida Research & Innovation Center, Port St. Lucie, FL, USA
| | - Michael Carlock
- Cleveland Clinic Florida Research & Innovation Center, Port St. Lucie, FL, USA
| | - Hyungwon Choi
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christine Vogel
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA.
| |
Collapse
|
24
|
Zhou Y, Chen H, Zhong W, Tao YJ. Collagen and actin network mediate antiviral immunity against Orsay virus in C. elegans intestinal cells. PLoS Pathog 2024; 20:e1011366. [PMID: 38190406 PMCID: PMC10798621 DOI: 10.1371/journal.ppat.1011366] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 01/19/2024] [Accepted: 12/18/2023] [Indexed: 01/10/2024] Open
Abstract
C. elegans is a free-living nematode that is widely used as a small animal model for studying fundamental biological processes and disease mechanisms. Since the discovery of the Orsay virus in 2011, C. elegans also holds the promise of dissecting virus-host interaction networks and innate antiviral immunity pathways in an intact animal. Orsay virus primarily targets the worm intestine, causing enlarged intestinal lumen as well as visible changes to infected cells such as liquefaction of cytoplasm and convoluted apical border. Previous studies of Orsay virus identified that C. elegans is able to mount antiviral responses by DRH-1/RIG-I mediated RNA interference and Intracellular Pathogen Response, a uridylyltransferase that destabilizes viral RNAs by 3' end uridylation, and ubiquitin protein modifications and turnover. To comprehensively search for novel antiviral pathways in C. elegans, we performed genome-wide RNAi screens by bacterial feeding using existing bacterial RNAi libraries covering 94% of the entire genome. Out of the 106 potential antiviral gene hits identified, we investigated those in three new pathways: collagens, actin remodelers, and epigenetic regulators. By characterizing Orsay virus infection in RNAi and mutant worms, our results indicate that collagens likely form a physical barrier in intestine cells to inhibit viral infection by preventing Orsay virus entry. Furthermore, evidence suggests that actin remodeling proteins (unc-34, wve-1 and wsp-1) and chromatin remodelers (nurf-1 and isw-1) exert their antiviral activities by regulating the intestinal actin (act-5), a critical component of the terminal web which likely function as another physical barrier to prevent Orsay infection.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| | - Hanqiao Chen
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| | - Weiwei Zhong
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| | - Yizhi Jane Tao
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| |
Collapse
|
25
|
Yan L, Wang P, Zhao C, Zhang B, Zhang B, Guo J, Qiu L. Development of a spotted sea bass (Lateolabrax maculatus) bulbus arteriosus cell line and its application to fish virology and immunology. FISH & SHELLFISH IMMUNOLOGY 2024; 144:109298. [PMID: 38122954 DOI: 10.1016/j.fsi.2023.109298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/05/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
The bulbus arteriosus tissue of teleosts, which is located at the forefront of the heart, is used to reduce the pulse pressure. In this study, we constructed a permanent cell line (LmAB) for the first time using bulbus arteriosus tissue from spotted sea bass (Lateolabrax maculatus). This cell line has been passaged more than 80 times. Currently, it can be subcultured in L-15 medium with 8 % fetal bovine serum added. The optimal fetal bovine serum concentration and culture temperature for LmAB cells at 62 passages are 20 % and 28 °C, respectively. This cell line consists predominantly of epithelial-like cells. We used 18S rRNA gene sequencing to confirm that LmAB cells originated from spotted sea bass. Karyotype analysis revealed that 43 % of LmAB cells in passage 63 had 48 chromosomes. Exogenous plasmid transfection revealed that LmAB cells can express the green fluorescent protein gene with a transfection efficiency of up to 40 %, indicating that these cells can be used for in vitro genetic research. LmAB cells showed susceptibility to nervous necrosis virus, largemouth bass ulcer syndrome virus, and infectious spleen and kidney necrosis virus, which results in severe cytopathic effects. PCR analysis verified that these viruses can replicate in LmAB cells, and analysis of cytoskeletal F-actin patterns verified that infected cells exhibit serious changes in their actin cytoskeleton. LmAB cells infected with these three viruses showed increased expressions of interferon signaling pathway genes (IFNd, IFNγ-rel, and ISG15), indicating that the host interferon signaling pathway participates in the antiviral immune response. These findings indicate that our newly developed LmAB cell line is a valuable resource for future research in genetics, virology, and immunology.
Collapse
Affiliation(s)
- Lulu Yan
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Sanya Tropical Fisheries Research Institute, Sanya, China
| | - Pengfei Wang
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Sanya Tropical Fisheries Research Institute, Sanya, China
| | - Chao Zhao
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Sanya Tropical Fisheries Research Institute, Sanya, China
| | - Bo Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Sanya Tropical Fisheries Research Institute, Sanya, China
| | - Bo Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Sanya Tropical Fisheries Research Institute, Sanya, China
| | - Jieyun Guo
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Lihua Qiu
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China; Sanya Tropical Fisheries Research Institute, Sanya, China; Key Laboratory of Aquatic Genomics, Ministry of Agriculture and Rural Affairs, Chinese Academy of Fishery Science, Beijing, China.
| |
Collapse
|
26
|
Artcibasova A, Wang L, Anchisi S, Yamauchi Y, Schmolke M, Matthias P, Stelling J. A quantitative model for virus uncoating predicts influenza A infectivity. Cell Rep 2023; 42:113558. [PMID: 38103200 DOI: 10.1016/j.celrep.2023.113558] [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: 06/30/2022] [Revised: 10/13/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
For virus infection of new host cells, the disassembly of the protective outer protein shell (capsid) is a critical step, but the mechanisms and host-virus interactions underlying the dynamic, active, and regulated uncoating process are largely unknown. Here, we develop an experimentally supported, multiscale kinetics model that elucidates mechanisms of influenza A virus (IAV) uncoating in cells. Biophysical modeling demonstrates that interactions between capsid M1 proteins, host histone deacetylase 6 (HDAC6), and molecular motors can physically break the capsid in a tug-of-war mechanism. Biochemical analysis and biochemical-biophysical modeling identify unanchored ubiquitin chains as essential and allow robust prediction of uncoating efficiency in cells. Remarkably, the different infectivity of two clinical strains can be ascribed to a single amino acid variation in M1 that affects binding to HDAC6. By identifying crucial modules of viral infection kinetics, the mechanisms and models presented here could help formulate novel strategies for broad-range antiviral treatment.
Collapse
Affiliation(s)
- Alina Artcibasova
- Department of Biosystems Science and Engineering and SIB Swiss Institute of Bioinformatics, ETH Zurich, 4058 Basel, Switzerland
| | - Longlong Wang
- Friedrich Miescher Institute for Biomedical Research (FMI), 4058 Basel, Switzerland; Faculty of Sciences, University of Basel, 4031 Basel, Switzerland
| | - Stephanie Anchisi
- Department of Microbiology and Molecular Medicine and Geneva Center of Inflammation Research, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Yohei Yamauchi
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Mirco Schmolke
- Department of Microbiology and Molecular Medicine and Geneva Center of Inflammation Research, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Patrick Matthias
- Friedrich Miescher Institute for Biomedical Research (FMI), 4058 Basel, Switzerland; Faculty of Sciences, University of Basel, 4031 Basel, Switzerland.
| | - Jörg Stelling
- Department of Biosystems Science and Engineering and SIB Swiss Institute of Bioinformatics, ETH Zurich, 4058 Basel, Switzerland.
| |
Collapse
|
27
|
Inamoto T, Furuta K, Han C, Uneme M, Kano T, Ishikawa K, Kaito C. Short-chain fatty acids stimulate dendrite elongation in dendritic cells by inhibiting histone deacetylase. FEBS J 2023; 290:5794-5810. [PMID: 37646105 DOI: 10.1111/febs.16945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/08/2023] [Accepted: 08/29/2023] [Indexed: 09/01/2023]
Abstract
Dendritic cells activate immune responses by presenting pathogen-derived molecules. The dendrites of dendritic cells contribute to the incorporation of foreign antigens or presenting antigens to T cells. Short-chain fatty acids (SCFAs), such as acetic, propionic, butyric and valeric acids, have many effects on immune responses by activating specific receptors or inhibiting a histone deacetylase (HDAC), although their effect on dendrite formation in dendritic cells is unknown. In the present study, we aimed to investigate the effect of SCFAs on dendrite elongation using a dendritic cell line (DC2.4 cells) and mouse bone marrow-derived dendritic cells. We found that SCFAs induced dendrite elongation. The elongation was reduced by inhibitors of Src family kinase (SFK), phosphatidylinositol-3 kinase (PI3K), Rho family GTPases (Cdc42, Rac1) or actin polymerization, indicating that SCFAs promote dendrite elongation by activating actin polymerization via the SFK/PI3K/Rho family GTPase signaling pathway. We showed that agonists for SCFA receptors GPR43 and GPR109a did not promote dendrite elongation. By contrast, HDAC inhibitors, including trichostatin A, promoted dendrite elongation in DC2.4 cells, and the promoting activity of trichostatin A was decreased by inhibiting the SFK/PI3K/Rho family GTPase signaling pathway or actin polymerization. Furthermore, DC2.4 cells treated with valeric acid showed enhanced uptake of soluble proteins, insoluble beads and Staphylococcus aureus. We also found that treatment with valeric acid enhanced major histocompatibility complex class II-mediated antigen presentation in bone marrow-derived dendritic cells. These results suggest that SCFAs promote dendrite elongation by inhibiting HDAC, stimulating the SFK/PI3K/Rho family pathway and activating actin polymerization, resulting in increased antigen uptake and presentation in dendritic cells.
Collapse
Affiliation(s)
- Takuho Inamoto
- Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Japan
| | - Kazuyuki Furuta
- Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Japan
| | - Cheng Han
- Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Japan
| | - Mio Uneme
- Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Japan
| | - Tomonori Kano
- Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Japan
| | - Kazuya Ishikawa
- Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Japan
| | - Chikara Kaito
- Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Japan
| |
Collapse
|
28
|
Halász H, Szatmári Z, Kovács K, Koppán M, Papp S, Szabó-Meleg E, Szatmári D. Changes of Ex Vivo Cervical Epithelial Cells Due to Electroporation with JMY. Int J Mol Sci 2023; 24:16863. [PMID: 38069185 PMCID: PMC10706833 DOI: 10.3390/ijms242316863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/20/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
The ionic environment within the nucleoplasm might diverge from the conditions found in the cytoplasm, potentially playing a role in the cellular stress response. As a result, it is conceivable that interactions of nuclear actin and actin-binding proteins (ABPs) with apoptosis factors may differ in the nucleoplasm and cytoplasm. The primary intracellular stress response is Ca2+ influx. The junctional mediating and regulating Y protein (JMY) is an actin-binding protein and has the capability to interact with the apoptosis factor p53 in a Ca2+-dependent manner, forming complexes that play a regulatory role in cytoskeletal remodelling and motility. JMY's presence is observed in both the cytoplasm and nucleoplasm. Here, we show that ex vivo ectocervical squamous cells subjected to electroporation with JMY protein exhibited varying morphological alterations. Specifically, the highly differentiated superficial and intermediate cells displayed reduced nuclear size. In inflamed samples, nuclear enlargement and simultaneous cytoplasmic reduction were observable and showed signs of apoptotic processes. In contrast, the less differentiated parabasal and metaplastic cells showed increased cytoplasmic activity and the formation of membrane protrusions. Surprisingly, in severe inflammation, vaginosis or ASC-US (Atypical Squamous Cells of Undetermined Significance), JMY appears to influence only the nuclear and perinuclear irregularities of differentiated cells, and cytoplasmic abnormalities still existed after the electroporation. Our observations can provide an appropriate basis for the exploration of the relationship between cytopathologically relevant morphological changes of epithelial cells and the function of ABPs. This is particularly important since ABPs are considered potential diagnostic and therapeutic biomarkers for both cancers and chronic inflammation.
Collapse
Affiliation(s)
- Henriett Halász
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (H.H.); (E.S.-M.)
| | | | - Krisztina Kovács
- Department of Pathology, Medical School, University of Pécs, 7624 Pécs, Hungary;
| | | | - Szilárd Papp
- DaVinci Clinics, 7635 Pécs, Hungary; (M.K.); (S.P.)
| | - Edina Szabó-Meleg
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (H.H.); (E.S.-M.)
| | - Dávid Szatmári
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (H.H.); (E.S.-M.)
| |
Collapse
|
29
|
Sasivimolrattana T, Bhattarakosol P. Impact of actin polymerization and filopodia formation on herpes simplex virus entry in epithelial, neuronal, and T lymphocyte cells. Front Cell Infect Microbiol 2023; 13:1301859. [PMID: 38076455 PMCID: PMC10704452 DOI: 10.3389/fcimb.2023.1301859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
Herpes simplex virus type 1 (HSV-1) has been known as a common viral pathogen that can infect several parts of the body, leading to various clinical manifestations. According to this diverse manifestation, HSV-1 infection in many cell types was demonstrated. Besides the HSV-1 cell tropism, e.g., fibroblast, epithelial, mucosal cells, and neurons, HSV-1 infections can occur in human T lymphocyte cells, especially in activated T cells. In addition, several studies found that actin polymerization and filopodia formation support HSV-1 infection in diverse cell types. Hence, the goal of this review is to explore the mechanism of HSV-1 infection in various types of cells involving filopodia formation and highlight potential future directions for HSV-1 entry-related research. Moreover, this review covers several strategies for possible anti-HSV drugs focused on the entry step, offering insights into potential therapeutic interventions.
Collapse
Affiliation(s)
| | - Parvapan Bhattarakosol
- Center of Excellence in Applied Medical Virology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Division of Virology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
30
|
Zhang Y, Zhang X, Li Z, Zhao W, Yang H, Zhao S, Tang D, Zhang Q, Li Z, Liu H, Li H, Li B, Lappalainen P, Xu T, Cui Z, Jiu Y. Single particle tracking reveals SARS-CoV-2 regulating and utilizing dynamic filopodia for viral invasion. Sci Bull (Beijing) 2023; 68:2210-2224. [PMID: 37661543 DOI: 10.1016/j.scib.2023.08.031] [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: 03/24/2023] [Revised: 06/22/2023] [Accepted: 08/11/2023] [Indexed: 09/05/2023]
Abstract
Although severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) entry mechanism has been explored, little is known about how SARS-CoV-2 regulates the subcellular structural remodeling to invade multiple organs and cell types. Here, we unveil how SARS-CoV-2 boosts and utilizes filopodia to enter the target cells by real-time imaging. Using SARS-CoV-2 single virus-like particle (VLP) tracking in live cells and sparse deconvolution algorithm, we uncover that VLPs utilize filopodia to reach the entry site in two patterns, "surfing" and "grabbing", which avoid the virus from randomly searching on the plasma membrane. Moreover, combining mechanical simulation, we elucidate that the formation of virus-induced filopodia and the retraction speed of filopodia depend on cytoskeleton dynamics and friction resistance at the substrate surface caused by loading-virus gravity, respectively. Further, we discover that the entry process of SARS-CoV-2 via filopodia depends on Cdc42 activity and actin-associated proteins fascin, formin, and Arp2/3. Together, our results highlight that the spatial-temporal regulation of actin cytoskeleton by SARS-CoV-2 infection makes filopodia as a highway for virus entry and potentiates it as an antiviral target.
Collapse
Affiliation(s)
- Yue Zhang
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaowei Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Zhongyi Li
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Weisong Zhao
- Innovation Photonics and Imaging Center, School of Instrumentation Science and Engineering, Harbin Institute of Technology, Harbin 150080, China
| | - Hui Yang
- University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Shuangshuang Zhao
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Daijiao Tang
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Zhang
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zonghong Li
- Guangzhou Laboratory, Guangzhou 510005, China
| | | | - Haoyu Li
- Innovation Photonics and Imaging Center, School of Instrumentation Science and Engineering, Harbin Institute of Technology, Harbin 150080, China
| | - Bo Li
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Pekka Lappalainen
- Institute of Biotechnology and Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | - Tao Xu
- Guangzhou Laboratory, Guangzhou 510005, China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Yaming Jiu
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
31
|
Sajewicz-Krukowska J, Mirosław P, Jastrzębski JP, Domańska-Blicharz K, Tarasiuk K, Marzec-Kotarska B. miRNA Expression Signatures Induced by Chicken Astrovirus Infection in Chickens. Int J Mol Sci 2023; 24:15128. [PMID: 37894809 PMCID: PMC10606767 DOI: 10.3390/ijms242015128] [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: 09/12/2023] [Revised: 09/27/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
miRNAs represent ubiquitous regulators of gene expression and play an important and pivotal regulatory role in viral disease pathogenesis and virus-host interactions. Although previous studies have provided basic data for understanding the role of miRNAs in the molecular mechanisms of viral infection in birds, the role of miRNAs in the regulation of host responses to chicken astrovirus (CAstV) infection in chickens is not yet understood. In our study, we applied next-generation sequencing to profile miRNA expression in CAstV-infected chickens and to decipher miRNA-targeted specific signaling pathways engaged in potentially vital virus-infection biological processes. Among the 1354 detected miRNAs, we identified 58 mature miRNAs that were significantly differentially expressed in infected birds. Target prediction resulted in 4741 target genes. GO and KEGG pathway enrichment analyses showed that the target genes were mainly involved in the regulation of cellular processes and immune responses.
Collapse
Affiliation(s)
- Joanna Sajewicz-Krukowska
- Department of Poultry Diseases, National Veterinary Research Institute, 24-100 Puławy, Poland; (K.D.-B.); (K.T.)
| | - Paweł Mirosław
- Foundation of Research and Science Development, 01-242 Warsaw, Poland;
| | - Jan P. Jastrzębski
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Katarzyna Domańska-Blicharz
- Department of Poultry Diseases, National Veterinary Research Institute, 24-100 Puławy, Poland; (K.D.-B.); (K.T.)
| | - Karolina Tarasiuk
- Department of Poultry Diseases, National Veterinary Research Institute, 24-100 Puławy, Poland; (K.D.-B.); (K.T.)
| | - Barbara Marzec-Kotarska
- Department of Clinical Pathomorphology, The Medical University of Lublin, 20-090 Lublin, Poland;
| |
Collapse
|
32
|
Dabrowska A, Botwina P, Barreto-Duran E, Kubisiak A, Obloza M, Synowiec A, Szczepanski A, Targosz-Korecka M, Szczubialka K, Nowakowska M, Pyrc K. Reversible rearrangement of the cellular cytoskeleton: A key to the broad-spectrum antiviral activity of novel amphiphilic polymers. Mater Today Bio 2023; 22:100763. [PMID: 37600352 PMCID: PMC10433002 DOI: 10.1016/j.mtbio.2023.100763] [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: 05/24/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/22/2023] Open
Abstract
The battle against emerging viral infections has been uneven, as there is currently no broad-spectrum drug available to contain the spread of novel pathogens throughout the population. Consequently, the pandemic outbreak that occurred in early 2020 laid bare the almost empty state of the pandemic box. Therefore, the development of novel treatments with broad specificity has become a paramount concern in this post-pandemic era. Here, we propose copolymers of poly (sodium 2-(acrylamido)-2-methyl-1-propanesulfonate) (PAMPS) and poly (sodium 11-(acrylamido)undecanoate (AaU), both block (PAMPS75-b-PAaUn) and random (P(AMPSm-co-AaUn)) that show efficacy against a broad range of alpha and betacoronaviruses. Owing to their intricate architecture, these polymers exhibit a highly distinctive mode of action, modulating nano-mechanical properties of cells and thereby influencing viral replication. Through the employment of confocal and atomic force microscopy techniques, we discerned perturbations in actin and vimentin filaments, which correlated with modification of cellular elasticity and reduction of glycocalyx layer. Intriguingly, this process was reversible upon polymer removal from the cells. To ascertain the applicability of our findings, we assessed the efficacy and underlying mechanism of the inhibitors using fully differentiated human airway epithelial cultures, wherein near-complete abrogation of viral replication was documented. Given their mode of action, these polymers can be classified as biologically active nanomaterials that exploit a highly conserved molecular target-cellular plasticity-proffering the potential for truly broad-spectrum activity while concurrently for drug resistance development is minimal.
Collapse
Affiliation(s)
- Agnieszka Dabrowska
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387, Cracow, Poland
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Cracow, Poland
| | - Pawel Botwina
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387, Cracow, Poland
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Cracow, Poland
| | - Emilia Barreto-Duran
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387, Cracow, Poland
| | - Agata Kubisiak
- Department of Physics of Nanostructures and Nanotechnology, Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Jagiellonian University, Lojasiewicza 11, 30-348, Cracow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Lojasiewicza 11, 30-348, Cracow, Poland
| | - Magdalena Obloza
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Cracow, Poland
| | - Aleksandra Synowiec
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387, Cracow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Lojasiewicza 11, 30-348, Cracow, Poland
| | - Artur Szczepanski
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387, Cracow, Poland
| | - Marta Targosz-Korecka
- Department of Physics of Nanostructures and Nanotechnology, Faculty of Physics, Astronomy and Applied Computer Science, M. Smoluchowski Institute of Physics, Jagiellonian University, Lojasiewicza 11, 30-348, Cracow, Poland
| | - Krzysztof Szczubialka
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Cracow, Poland
| | - Maria Nowakowska
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Cracow, Poland
| | - Krzysztof Pyrc
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30-387, Cracow, Poland
| |
Collapse
|
33
|
Moniruzzaman M, Erazo Garcia MP, Farzad R, Ha AD, Jivaji A, Karki S, Sheyn U, Stanton J, Minch B, Stephens D, Hancks DC, Rodrigues RAL, Abrahao JS, Vardi A, Aylward FO. Virologs, viral mimicry, and virocell metabolism: the expanding scale of cellular functions encoded in the complex genomes of giant viruses. FEMS Microbiol Rev 2023; 47:fuad053. [PMID: 37740576 PMCID: PMC10583209 DOI: 10.1093/femsre/fuad053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/29/2023] [Accepted: 09/21/2023] [Indexed: 09/24/2023] Open
Abstract
The phylum Nucleocytoviricota includes the largest and most complex viruses known. These "giant viruses" have a long evolutionary history that dates back to the early diversification of eukaryotes, and over time they have evolved elaborate strategies for manipulating the physiology of their hosts during infection. One of the most captivating of these mechanisms involves the use of genes acquired from the host-referred to here as viral homologs or "virologs"-as a means of promoting viral propagation. The best-known examples of these are involved in mimicry, in which viral machinery "imitates" immunomodulatory elements in the vertebrate defense system. But recent findings have highlighted a vast and rapidly expanding array of other virologs that include many genes not typically found in viruses, such as those involved in translation, central carbon metabolism, cytoskeletal structure, nutrient transport, vesicular trafficking, and light harvesting. Unraveling the roles of virologs during infection as well as the evolutionary pathways through which complex functional repertoires are acquired by viruses are important frontiers at the forefront of giant virus research.
Collapse
Affiliation(s)
- Mohammad Moniruzzaman
- Rosenstiel School of Marine Atmospheric, and Earth Science, University of Miami, Coral Gables, FL 33149, United States
| | - Maria Paula Erazo Garcia
- Department of Biological Sciences, Virginia Tech, 926 West Campus Drive, Blacksburg, VA 24061, United States
| | - Roxanna Farzad
- Department of Biological Sciences, Virginia Tech, 926 West Campus Drive, Blacksburg, VA 24061, United States
| | - Anh D Ha
- Department of Biological Sciences, Virginia Tech, 926 West Campus Drive, Blacksburg, VA 24061, United States
| | - Abdeali Jivaji
- Department of Biological Sciences, Virginia Tech, 926 West Campus Drive, Blacksburg, VA 24061, United States
| | - Sangita Karki
- Department of Biological Sciences, Virginia Tech, 926 West Campus Drive, Blacksburg, VA 24061, United States
| | - Uri Sheyn
- Department of Biological Sciences, Virginia Tech, 926 West Campus Drive, Blacksburg, VA 24061, United States
| | - Joshua Stanton
- Department of Biological Sciences, Virginia Tech, 926 West Campus Drive, Blacksburg, VA 24061, United States
| | - Benjamin Minch
- Rosenstiel School of Marine Atmospheric, and Earth Science, University of Miami, Coral Gables, FL 33149, United States
| | - Danae Stephens
- Rosenstiel School of Marine Atmospheric, and Earth Science, University of Miami, Coral Gables, FL 33149, United States
| | - Dustin C Hancks
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, United States
| | - Rodrigo A L Rodrigues
- Laboratório de Vírus, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Jonatas S Abrahao
- Laboratório de Vírus, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Assaf Vardi
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Frank O Aylward
- Department of Biological Sciences, Virginia Tech, 926 West Campus Drive, Blacksburg, VA 24061, United States
- Center for Emerging, Zoonotic, and Arthropod-Borne Infectious Disease, Virginia Tech, Blacksburg, VA 24061, United States
| |
Collapse
|
34
|
Banik A, Ahmed SR, Shahid SB, Ahmed T, Tamanna HK, Marma H. Therapeutic Promises of Plant Metabolites against Monkeypox Virus: An In Silico Study. Adv Virol 2023; 2023:9919776. [PMID: 37693295 PMCID: PMC10492655 DOI: 10.1155/2023/9919776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
The monkeypox virus was still spreading in May 2022, with the first case identified in a person with travel ties to Nigeria. Using molecular docking-based techniques, we evaluated the efficiency of different bioactive chemicals obtained from plants against the monkeypox virus. A total of 56 plant compounds were evaluated for antimonekypox capabilities, with the top four candidates having a higher binding affinity than the control. We targeted the monkeypox profilin-like protein, which plays a key role in viral replication and assembly. Among the metabolites, curcumin showed the strongest binding affinity with a value of -37.43 kcal/mol, followed by gedunin (-34.89 kcal/mol), piperine (-34.58 kcal/mol), and coumadin (-34.14 kcal/mol). Based on ADME and toxicity assessments, the top four substances had no negative impacts. Furthermore, four compounds demonstrated resistance to deformability, which was corroborated by normal mode analysis. According to the bioactivity prediction study, the top compound target class was an enzyme, membrane receptor, and oxidoreductase. Furthermore, the study discovered that wortmannin, a gedunin analogue, can behave as an orthopoxvirus. The study found that these bioactive natural drug candidates could potentially work as monkeypox virus inhibitors. We recommended further experimental validation to confirm the promising findings of the study.
Collapse
Affiliation(s)
- Anik Banik
- Department of Plant and Environmental Biotechnology, Sylhet Agricultural University, Sylhet 3100, Bangladesh
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Sheikh Rashel Ahmed
- Department of Plant and Environmental Biotechnology, Sylhet Agricultural University, Sylhet 3100, Bangladesh
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Sonia Binte Shahid
- Department of Plant and Environmental Biotechnology, Sylhet Agricultural University, Sylhet 3100, Bangladesh
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Tufayel Ahmed
- Department of Plant and Environmental Biotechnology, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | | | - Hlamrasong Marma
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| |
Collapse
|
35
|
Yeung-Luk BH, Narayanan GA, Ghosh B, Wally A, Lee E, Mokaya M, Wankhade E, Zhang R, Lee B, Park B, Resnick J, Jedlicka A, Dziedzic A, Ramanathan M, Biswal S, Pekosz A, Sidhaye VK. SARS-CoV-2 infection alters mitochondrial and cytoskeletal function in human respiratory epithelial cells mediated by expression of spike protein. mBio 2023; 14:e0082023. [PMID: 37504520 PMCID: PMC10470579 DOI: 10.1128/mbio.00820-23] [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: 03/31/2023] [Accepted: 06/10/2023] [Indexed: 07/29/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, SCV2), which has resulted in higher morbidity and mortality rate than other respiratory viral infections, such as Influenza A virus (IAV) infection. Investigating the molecular mechanisms of SCV2-host infection vs IAV is vital in exploring antiviral drug targets against SCV2. We assessed differential gene expression in human nasal cells upon SCV2 or IAV infection using RNA sequencing. Compared to IAV, we observed alterations in both metabolic and cytoskeletal pathways suggestive of epithelial remodeling in the SCV2-infected cells, reminiscent of pathways activated as a response to chronic injury. We found that spike protein interaction with the epithelium was sufficient to instigate these epithelial responses using a SCV2 spike pseudovirus. Specifically, we found downregulation of the mitochondrial markers SIRT3 and TOMM22. Moreover, SCV2 spike infection increased extracellular acidification and decreased oxygen consumption rate in the epithelium. In addition, we observed cytoskeletal rearrangements with a reduction in the actin-severing protein cofilin-1 and an increase in polymerized actin, indicating epithelial cytoskeletal rearrangements. This study revealed distinct epithelial responses to SCV2 infection, with early mitochondrial dysfunction in the host cells and evidence of cytoskeletal remodeling that could contribute to the worsened outcome in COVID-19 patients compared to IAV patients. These changes in cell structure and energetics could contribute to cellular resilience early during infection, allowing for prolonged cell survival and potentially paving the way for more chronic symptoms. IMPORTANCE COVID-19 has caused a global pandemic affecting millions of people worldwide, resulting in a higher mortality rate and concerns of more persistent symptoms compared to influenza A. To study this, we compare lung epithelial responses to both viruses. Interestingly, we found that in response to SARS-CoV-2 infection, the cellular energetics changed and there were cell structural rearrangements. These changes in cell structure could lead to prolonged epithelial cell survival, even in the face of not working well, potentially contributing to the development of chronic symptoms. In summary, these findings represent strategies utilized by the cell to survive the infection but result in a fundamental shift in the epithelial phenotype, with potential long-term consequences, which could set the stage for the development of chronic lung disease or long COVID-19.
Collapse
Affiliation(s)
- Bonnie H. Yeung-Luk
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | | | - Baishakhi Ghosh
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Ara Wally
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Esther Lee
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Michelle Mokaya
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Esha Wankhade
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Rachel Zhang
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Brianna Lee
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Bongsoo Park
- Epigenetics and Stem Cell Aging, National Institute of Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Jessica Resnick
- W. Harry Feinstone Department of Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Anne Jedlicka
- W. Harry Feinstone Department of Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Amanda Dziedzic
- W. Harry Feinstone Department of Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Murugappan Ramanathan
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Outpatient Center, Baltimore, Maryland, USA
| | - Shyam Biswal
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Venkataramana K. Sidhaye
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
36
|
Inomura K, Pierella Karlusich JJ, Dutkiewicz S, Deutsch C, Harrison PJ, Bowler C. High Growth Rate of Diatoms Explained by Reduced Carbon Requirement and Low Energy Cost of Silica Deposition. Microbiol Spectr 2023; 11:e0331122. [PMID: 37010412 PMCID: PMC10269801 DOI: 10.1128/spectrum.03311-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 03/02/2023] [Indexed: 04/04/2023] Open
Abstract
The rapid growth of diatoms makes them one of the most pervasive and productive types of plankton in the world's ocean, but the physiological basis for their high growth rates remains poorly understood. Here, we evaluate the factors that elevate diatom growth rates, relative to other plankton, using a steady-state metabolic flux model that computes the photosynthetic C source from intracellular light attenuation and the carbon cost of growth from empirical cell C quotas, across a wide range of cell sizes. For both diatoms and other phytoplankton, growth rates decline with increased cell volume, consistent with observations, because the C cost of division increases with size faster than photosynthesis. However, the model predicts overall higher growth rates for diatoms due to reduced C requirements and the low energetic cost of Si deposition. The C savings from the silica frustule are supported by metatranscriptomic data from Tara Oceans, which show that the abundance of transcripts for cytoskeleton components in diatoms is lower than in other phytoplankton. Our results highlight the importance of understanding the origins of phylogenetic differences in cellular C quotas, and suggest that the evolution of silica frustules may play a critical role in the global dominance of marine diatoms. IMPORTANCE This study addresses a longstanding issue regarding diatoms, namely, their fast growth. Diatoms, which broadly are phytoplankton with silica frustules, are the world's most productive microorganisms and dominate in polar and upwelling regions. Their dominance is largely supported by their high growth rate, but the physiological reasoning behind that characteristic has been obscure. In this study, we combine a quantitative model and metatranscriptomic approaches and show that diatoms' low carbon requirements and low energy costs for silica frustule production are the key factors supporting their fast growth. Our study suggests that the effective use of energy-efficient silica as a cellular structure, instead of carbon, enables diatoms to be the most productive organisms in the global ocean.
Collapse
Affiliation(s)
- Keisuke Inomura
- Graduate School of Oceanography, University of Rhode Island, Narragansett, Rhode Island, USA
| | - Juan José Pierella Karlusich
- Institut de Biologie de l'Ecole Normale Supérieure, CNRS, INSERM, Université Paris Sciences et Lettres, Paris, France
- Faculty of Arts and Sciences, Division of Science, Harvard University, Cambridge, Massachusetts, USA
| | - Stephanie Dutkiewicz
- Department of Earth, Atmospheric, and Planetary Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Curtis Deutsch
- Department of Geosciences, Princeton University, Princeton, New Jersey, USA
| | - Paul J. Harrison
- Department of Earth, Ocean, and Atmospheric Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Chris Bowler
- Institut de Biologie de l'Ecole Normale Supérieure, CNRS, INSERM, Université Paris Sciences et Lettres, Paris, France
| |
Collapse
|
37
|
Liang M, Li Y, Zhang K, Zhu Y, Liang J, Liu M, Zhang S, Chen D, Liang H, Liang L, An S, Zhu X, He Z. Host factor DUSP5 potently inhibits dengue virus infection by modulating cytoskeleton rearrangement. Antiviral Res 2023; 215:105622. [PMID: 37149044 DOI: 10.1016/j.antiviral.2023.105622] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/08/2023]
Abstract
Cytoskeleton has been reported to play an essential role in facilitating the viral life cycle. However, whether the host can exert its antiviral effects by modulating the cytoskeleton is not fully understood. In this study, we identified that host factor DUSP5 was upregulated after dengue virus (DENV) infection. In addition, we demonstrated that overexpression of DUSP5 remarkably inhibited DENV replication. Conversely, the depletion of DUSP5 led to an increase in viral replication. Moreover, DUSP5 was found to restrain viral entry into host cells by suppressing F-actin rearrangement via negatively regulating the ERK-MLCK-Myosin IIB signaling axis. Depletion of dephosphorylase activity of DUSP5 abolished its above inhibitory effects. Furthermore, we also revealed that DUSP5 exhibited broad-spectrum antiviral effects against DENV and Zika virus. Taken together, our studies identified DUSP5 as a key host defense factor against viral infection and uncovered an intriguing mechanism by which the host exerts its antiviral effects through targeting cytoskeleton rearrangement.
Collapse
Affiliation(s)
- Minqi Liang
- School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yizhe Li
- School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Kexin Zhang
- School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yujia Zhu
- School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jingyao Liang
- School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Minjie Liu
- School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shuqing Zhang
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Delin Chen
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hao Liang
- Cancer Institute, Southern Medical University, Guangzhou, 510515, China
| | - Linyue Liang
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shu An
- Department of Laboratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China.
| | - Xun Zhu
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China; Central Laboratory, The Third People's Hospital of Zhuhai, Zhuhai, 519060, China.
| | - Zhenjian He
- School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China.
| |
Collapse
|
38
|
Boulton S, Crupi MJF, Singh S, Carter-Timofte ME, Azad T, Organ BC, He X, Gill R, Neault S, Jamieson T, Dave J, Kurmasheva N, Austin B, Petryk J, Singaravelu R, Huang BZ, Franco N, Babu K, Parks RJ, Ilkow CS, Olagnier D, Bell JC. Inhibition of Exchange Proteins Directly Activated by cAMP (EPAC) as a Strategy for Broad-Spectrum Antiviral Development. J Biol Chem 2023; 299:104749. [PMID: 37100284 PMCID: PMC10124099 DOI: 10.1016/j.jbc.2023.104749] [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: 02/08/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 04/28/2023] Open
Abstract
The recent SARS-CoV-2 and mpox outbreaks have highlighted the need to expand our arsenal of broad-spectrum antiviral agents for future pandemic preparedness. Host-directed antivirals are an important tool to accomplish this as they typically offer protection against a broader range of viruses than direct-acting antivirals and have a lower susceptibility to viral mutations that cause drug resistance. In this study, we investigate the Exchange Protein Activated by cAMP (EPAC) as a target for broad-spectrum antiviral therapy. We find that the EPAC-selective inhibitor, ESI-09 provides robust protection against a variety of viruses, including SARS-CoV-2 and Vaccinia (VACV) - an orthopoxvirus from the same family as mpox. We show, using a series of immunofluorescence experiments, that ESI-09 remodels the actin cytoskeleton through Rac1/Cdc42 GTPases and the Arp2/3 complex, impairing internalization of viruses that use clathrin-mediated endocytosis (e.g. VSV) or micropinocytosis (e.g. VACV). Additionally, we find that ESI-09 disrupts syncytia formation and inhibits cell-to-cell transmission of viruses such as measles and VACV. When administered to immune-deficient mice in an intranasal challenge model, ESI-09 protects mice from lethal doses of VACV and prevents formation of pox lesions. Altogether, our finding show that EPAC antagonists such as ESI-09 are promising candidates for broad-spectrum antiviral therapy that can aid in the fight against ongoing and future viral outbreaks.
Collapse
Affiliation(s)
- Stephen Boulton
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| | - Mathieu J F Crupi
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Siddharth Singh
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | | | - Taha Azad
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada; Faculty of Medicine and Health Sciences, Department of microbiology and infectious diseases, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada; Centre de Recherche du CHUS, Sherbrooke, QC J1H 5N4, Canada
| | - Bailey C Organ
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Xiaohong He
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Rida Gill
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Serge Neault
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Taylor Jamieson
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Jaahnavi Dave
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Naziia Kurmasheva
- Aarhus University, Department of Biomedicine, Aarhus C, 8000, Denmark
| | - Bradley Austin
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Julia Petryk
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Ragunath Singaravelu
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada; Public Health Agency of Canada, Ottawa, Ontario, Canada, K1A 0K9
| | - Ben Zhen Huang
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Noah Franco
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Kaaviya Babu
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Robin J Parks
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada; Department of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Carolina S Ilkow
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - David Olagnier
- Aarhus University, Department of Biomedicine, Aarhus C, 8000, Denmark
| | - John C Bell
- Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada; Department of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
39
|
Zhou Y, Zhong W, Tao YJ. Collagen and actin network mediate antiviral immunity against Orsay in C. elegans intestinal cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.20.537671. [PMID: 37131627 PMCID: PMC10153230 DOI: 10.1101/2023.04.20.537671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
C. elegans is a free-living nematode that is widely used as a small animal model for studying fundamental biological processes and disease mechanisms. Since the discovery of the Orsay virus in 2011, C. elegans also holds the promise of dissecting virus-host interaction networks and innate antiviral immunity pathways in an intact animal. Orsay primarily targets the worm intestine, causing enlarged intestinal lumen as well as visible changes to infected cells such as liquefaction of cytoplasm and rearrangement of the terminal web. Previous studies of Orsay identified that C. elegans is able to mount antiviral responses by DRH-1/RIG-I mediated RNA interference and Intracellular Pathogen Response, a uridylyltransferase that destabilizes viral RNAs by 3' end uridylation, and ubiquitin protein modifications and turnover. To comprehensively search for novel antiviral pathways in C. elegans, we performed genome-wide RNAi screens by bacterial feeding using existing bacterial RNAi libraries covering 94% of the entire genome. Out of the 106 antiviral genes identified, we investigated those in three new pathways: collagens, actin remodelers, and epigenetic regulators. By characterizing Orsay infection in RNAi and mutant worms, our results indicate that collagens likely form a physical barrier in intestine cells to inhibit viral infection by preventing Orsay entry. Furthermore, evidence suggests that the intestinal actin (act-5), which is regulated by actin remodeling proteins (unc-34, wve-1 and wsp-1), a Rho GTPase (cdc-42) and chromatin remodelers (nurf-1 and isw-1), also provides antiviral immunity against Orsay possibly through another physical barrier presented as the terminal web.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Biosciences, Rice University, MS-605, Houston, Texas, 77005, USA
| | - Weiwei Zhong
- Department of Biosciences, Rice University, MS-605, Houston, Texas, 77005, USA
| | - Yizhi Jane Tao
- Department of Biosciences, Rice University, MS-605, Houston, Texas, 77005, USA
| |
Collapse
|
40
|
Wyatt J, Chan YK, Hess M, Tavassoli M, Müller MM. Semisynthesis reveals apoptin as a tumour-selective protein prodrug that causes cytoskeletal collapse. Chem Sci 2023; 14:3881-3892. [PMID: 37035694 PMCID: PMC10074440 DOI: 10.1039/d2sc04481a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 03/15/2023] [Indexed: 03/18/2023] Open
Abstract
Apoptin is a small viral protein capable of inducing cell death selectively in cancer cells. Despite its potential as an anticancer agent, relatively little is known about its mechanism of toxicity and cancer-selectivity. Previous experiments suggest that cancer-selective phosphorylation modulates apoptin toxicity, although a lack of chemical tools has hampered the dissection of underlying mechanisms. Here, we describe structure-function studies with site-specifically phosphorylated apoptin (apoptin-T108ph) in living cells which revealed that Thr108 phosphorylation is the selectivity switch for apoptin toxicity. Mechanistic investigations link T108ph to actin binding, cytoskeletal disruption and downstream inhibition of anoikis-resistance as well as cancer cell invasion. These results establish apoptin as a protein pro-drug, selectively activated in cancer cells by phosphorylation, which disrupts the cytoskeleton and promotes cell death. We anticipate that this mechanism provides a framework for the design of next generation anticancer proteins with enhanced selectivity and potency.
Collapse
Affiliation(s)
- Jasmine Wyatt
- Department of Molecular Oncology, King's College London Guy's Hospital Campus, Hodgkin Building London SE1 1UL UK
- Department of Chemistry, King's College London Britannia House, 7 Trinity Street London SE1 1DB UK
| | - Yuen Ka Chan
- Department of Molecular Oncology, King's College London Guy's Hospital Campus, Hodgkin Building London SE1 1UL UK
| | - Mateusz Hess
- Department of Chemistry, King's College London Britannia House, 7 Trinity Street London SE1 1DB UK
| | - Mahvash Tavassoli
- Department of Molecular Oncology, King's College London Guy's Hospital Campus, Hodgkin Building London SE1 1UL UK
| | - Manuel M Müller
- Department of Chemistry, King's College London Britannia House, 7 Trinity Street London SE1 1DB UK
| |
Collapse
|
41
|
Talukdar SN, Osan J, Ryan K, Grove B, Perley D, Kumar BD, Yang S, Dallman S, Hollingsworth L, Bailey KL, Mehedi M. RSV-induced expanded ciliated cells contribute to bronchial wall thickening. Virus Res 2023; 327:199060. [PMID: 36746339 PMCID: PMC10007709 DOI: 10.1016/j.virusres.2023.199060] [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: 11/20/2022] [Revised: 01/02/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023]
Abstract
Viral infection, particularly respiratory syncytial virus (RSV), causes inflammation in the bronchiolar airways (bronchial wall thickening, also known as bronchiolitis). This bronchial wall thickening is a common pathophysiological feature in RSV infection, but it causes more fatalities in infants than in children and adults. However, the molecular mechanism of RSV-induced bronchial wall thickening remains unknown, particularly in healthy adults. Using highly differentiated pseudostratified airway epithelium generated from primary human bronchial epithelial cells, we revealed RSV-infects primarily ciliated cells. The infected ciliated cells expanded substantially without compromising epithelial membrane integrity and ciliary functions and contributed to the increased height of the airway epithelium. Furthermore, we identified multiple factors, e.g., cytoskeletal (ARP2/3-complex-driven actin polymerization), immunological (IP10/CXCL10), and viral (NS2), contributing to RSV-induced uneven epithelium height increase in vitro. Thus, RSV-infected expanded cells contribute to a noncanonical inflammatory phenotype, which contributes to bronchial wall thickening in the airway, and is termed cytoskeletal inflammation.
Collapse
Affiliation(s)
- Sattya N Talukdar
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Jaspreet Osan
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Ken Ryan
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Bryon Grove
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Danielle Perley
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Bony D Kumar
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Shirley Yang
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Sydney Dallman
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Lauren Hollingsworth
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States
| | - Kristina L Bailey
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep and Allergy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Masfique Mehedi
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND, United States.
| |
Collapse
|
42
|
Roa-Linares VC, Escudero-Flórez M, Vicente-Manzanares M, Gallego-Gómez JC. Host Cell Targets for Unconventional Antivirals against RNA Viruses. Viruses 2023; 15:v15030776. [PMID: 36992484 PMCID: PMC10058429 DOI: 10.3390/v15030776] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/12/2023] [Accepted: 02/28/2023] [Indexed: 03/31/2023] Open
Abstract
The recent COVID-19 crisis has highlighted the importance of RNA-based viruses. The most prominent members of this group are SARS-CoV-2 (coronavirus), HIV (human immunodeficiency virus), EBOV (Ebola virus), DENV (dengue virus), HCV (hepatitis C virus), ZIKV (Zika virus), CHIKV (chikungunya virus), and influenza A virus. With the exception of retroviruses which produce reverse transcriptase, the majority of RNA viruses encode RNA-dependent RNA polymerases which do not include molecular proofreading tools, underlying the high mutation capacity of these viruses as they multiply in the host cells. Together with their ability to manipulate the immune system of the host in different ways, their high mutation frequency poses a challenge to develop effective and durable vaccination and/or treatments. Consequently, the use of antiviral targeting agents, while an important part of the therapeutic strategy against infection, may lead to the selection of drug-resistant variants. The crucial role of the host cell replicative and processing machinery is essential for the replicative cycle of the viruses and has driven attention to the potential use of drugs directed to the host machinery as therapeutic alternatives to treat viral infections. In this review, we discuss small molecules with antiviral effects that target cellular factors in different steps of the infectious cycle of many RNA viruses. We emphasize the repurposing of FDA-approved drugs with broad-spectrum antiviral activity. Finally, we postulate that the ferruginol analog (18-(phthalimide-2-yl) ferruginol) is a potential host-targeted antiviral.
Collapse
Affiliation(s)
- Vicky C Roa-Linares
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| | - Manuela Escudero-Flórez
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain
| | - Juan C Gallego-Gómez
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| |
Collapse
|
43
|
Hussain H, Ganesh A, Milane L, Amiji M. Lessons learned from the SARS-CoV-2 pandemic; from nucleic acid nanomedicines, to clinical trials, herd immunity, and the vaccination divide. Expert Opin Drug Deliv 2023; 20:489-506. [PMID: 36890642 DOI: 10.1080/17425247.2023.2189697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
INTRODUCTION In November 2019, the idea of a zoonotic virus crossing over to human transmission in a seafood market in Wuhan, China, and then soaring across the globe to claim over 6.3 million lives and persisting to date, seemed more like wild science fiction than a future reality. As the SARS-CoV-2 pandemic continues, it is important to hallmark the imprints the pandemic has made on science. AREAS COVERED This review covers the biology of SARS-CoV-2, vaccine formulations and trials, the concept of 'herd resistance,' and the vaccination divide. EXPERT OPINION The SARS-CoV-2 pandemic has changed the landscape of medicine. The rapid approval of SARS-CoV-2 vaccines has changed the culture of drug development and clinical approvals. This change is already leading to more accelerated trials. The RNA vaccines have opened the market for nucleic acid therapies and the applications are limitless - from cancer to influenza. A phenomenon that has occurred is that the low efficacy of current vaccines and the rapid mutation rate of the virus is preventing herd immunity from being attained. Instead, herd resistance is being acquired. Even with future, more effective vaccines, anti-vaccination attitudes will continue to challenge the quest for SARS-CoV-2 herd immunity.
Collapse
Affiliation(s)
| | - Aishwarya Ganesh
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Lara Milane
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Mansoor Amiji
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| |
Collapse
|
44
|
Cellular Targets of HIV-1 Protease: Just the Tip of the Iceberg? Viruses 2023; 15:v15030712. [PMID: 36992421 PMCID: PMC10053624 DOI: 10.3390/v15030712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/05/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Human immunodeficiency virus 1 (HIV-1) viral protease (PR) is one of the most studied viral enzymes and a crucial antiviral target. Despite its well-characterized role in virion maturation, an increasing body of research is starting to focus on its ability to cleave host cell proteins. Such findings are apparently in contrast with the dogma of HIV-1 PR activity being restricted to the interior of nascent virions and suggest catalytic activity within the host cell environment. Given the limited amount of PR present in the virion at the time of infection, such events mainly occur during late viral gene expression, mediated by newly synthesized Gag-Pol polyprotein precursors, rather than before proviral integration. HIV-1 PR mainly targets proteins involved in three different processes: those involved in translation, those controlling cell survival, and restriction factors responsible for innate/intrinsic antiviral responses. Indeed, by cleaving host cell translation initiation factors, HIV-1 PR can impair cap-dependent translation, thus promoting IRES-mediated translation of late viral transcripts and viral production. By targeting several apoptotic factors, it modulates cell survival, thus promoting immune evasion and viral dissemination. Additionally, HIV-1 PR counteracts restriction factors incorporated in the virion that would otherwise interfere with nascent virus vitality. Thus, HIV-1 PR appears to modulate host cell function at different times and locations during its life cycle, thereby ensuring efficient viral persistency and propagation. However, we are far from having a complete picture of PR-mediated host cell modulation, which is emerging as a field that needs further investigation.
Collapse
|
45
|
Villalonga E, Mosrin C, Normand T, Girardin C, Serrano A, Žunar B, Doudeau M, Godin F, Bénédetti H, Vallée B. LIM Kinases, LIMK1 and LIMK2, Are Crucial Node Actors of the Cell Fate: Molecular to Pathological Features. Cells 2023; 12:cells12050805. [PMID: 36899941 PMCID: PMC10000741 DOI: 10.3390/cells12050805] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
LIM kinase 1 (LIMK1) and LIM kinase 2 (LIMK2) are serine/threonine and tyrosine kinases and the only two members of the LIM kinase family. They play a crucial role in the regulation of cytoskeleton dynamics by controlling actin filaments and microtubule turnover, especially through the phosphorylation of cofilin, an actin depolymerising factor. Thus, they are involved in many biological processes, such as cell cycle, cell migration, and neuronal differentiation. Consequently, they are also part of numerous pathological mechanisms, especially in cancer, where their involvement has been reported for a few years and has led to the development of a wide range of inhibitors. LIMK1 and LIMK2 are known to be part of the Rho family GTPase signal transduction pathways, but many more partners have been discovered over the decades, and both LIMKs are suspected to be part of an extended and various range of regulation pathways. In this review, we propose to consider the different molecular mechanisms involving LIM kinases and their associated signalling pathways, and to offer a better understanding of their variety of actions within the physiology and physiopathology of the cell.
Collapse
Affiliation(s)
- Elodie Villalonga
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Christine Mosrin
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Thierry Normand
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Caroline Girardin
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Amandine Serrano
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Bojan Žunar
- Laboratory for Biochemistry, Department of Chemistry and Biochemistry, Faculty of Food Technology and Biotechnology, University of Zagreb, 10000 Zagreb, Croatia
| | - Michel Doudeau
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Fabienne Godin
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Hélène Bénédetti
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
| | - Béatrice Vallée
- Centre de Biophysique Moléculaire; UPR4301, CNRS, University of Orleans and INSERM, CEDEX 2, 45071 Orleans, France
- Correspondence: ; Tel.: +33-(0)2-38-25-76-11
| |
Collapse
|
46
|
Astrocytes in the pathophysiology of neuroinfection. Essays Biochem 2023; 67:131-145. [PMID: 36562155 DOI: 10.1042/ebc20220082] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022]
Abstract
Key homeostasis providing cells in the central nervous system (CNS) are astrocytes, which belong to the class of cells known as atroglia, a highly heterogeneous type of neuroglia and a prominent element of the brain defence. Diseases evolve due to altered homeostatic state, associated with pathology-induced astroglia remodelling represented by reactive astrocytes, astroglial atrophy and astrodegeneration. These features are hallmarks of most infectious insults, mediated by bacteria, protozoa and viruses; they are also prominent in the systemic infection. The COVID-19 pandemic revived the focus into neurotropic viruses such as SARS-CoV2 (Coronaviridae) but also the Flaviviridae viruses including tick-borne encephalitis (TBEV) and Zika virus (ZIKV) causing the epidemic in South America prior to COVID-19. Astrocytes provide a key response to neurotropic infections in the CNS. Astrocytes form a parenchymal part of the blood-brain barrier, the site of virus entry into the CNS. Astrocytes exhibit aerobic glycolysis, a form of metabolism characteristic of highly morphologically plastic cells, like cancer cells, hence a suitable milieu for multiplication of infectious agent, including viral particles. However, why the protection afforded by astrocytes fails in some circumstances is an open question to be studied in the future.
Collapse
|
47
|
Gee YJ, Sea YL, Lal SK. Viral modulation of lipid rafts and their potential as putative antiviral targets. Rev Med Virol 2023; 33:e2413. [PMID: 36504273 DOI: 10.1002/rmv.2413] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 11/12/2022] [Accepted: 11/20/2022] [Indexed: 12/14/2022]
Abstract
Lipid rafts are ubiquitous in cells. They are identified as cholesterol and glycosphingolipid enriched microdomains on cellular membranes. They serve as platforms for cellular communications by functioning in signal transduction and membrane trafficking. Such structural organisation fulfils cellular needs for normal function, but at the same time increases vulnerability of cells to pathogen invasion. Viruses rely heavily on lipid rafts in basically every stage of the viral life cycle for successful infection. Various mechanisms of lipid rafts modification exploited by diverse viruses for attachment, internalisation, membrane fusion, genome replication, assembly and release have been brought to light. This review focuses on virus-raft interactions and how a wide range of viruses manipulate lipid rafts at distinct stages of infection. The importance of virus-raft interactions in viral infections has inspired researchers to discover and develop antivirals that target this interaction, such as statins, methyl-β-cyclodextrin, viperin, 25-hydroxycholesterol and even anti-malarial drugs. The therapeutic modulations of lipid rafts as potential antiviral intervention from in vitro and in vivo evidence are discussed herein.
Collapse
Affiliation(s)
- Yee Jing Gee
- School of Science, Monash University, Bandar Sunway, Selangor DE, Malaysia
| | - Yi Lin Sea
- School of Science, Monash University, Bandar Sunway, Selangor DE, Malaysia
| | - Sunil Kumar Lal
- School of Science, Monash University, Bandar Sunway, Selangor DE, Malaysia.,Tropical Medicine & Biology Platform, Monash University, Bandar Sunway, Selangor DE, Malaysia
| |
Collapse
|
48
|
Liu W, Shang X, Wen W, Ren X, Qin L, Li X, Qian P. Seneca Valley virus enters cells through multiple pathways and traffics intracellularly via the endolysosomal pathway. J Gen Virol 2023; 104. [PMID: 36947577 DOI: 10.1099/jgv.0.001833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
Seneca Valley virus (SVV, also known as Senecavirus A), an oncolytic virus, is a nonenveloped, positive-strand RNA virus and the sole member of the genus Senecavirus within the family Picornaviridae. The mechanisms of SVV entry into cells are currently almost unknown. In the present study, we found that SVV entry into HEK293T cells is acidic pH-dependent by using ammonium chloride (NH4Cl) and chloroquine, both of which could inhibit SVV infection. We confirmed that dynamin II is required for SVV entry by using dynasore, silencing the dynamin II protein, or expressing the dominant-negative (DN) K44A mutant of dynamin II. Then, we discovered that chlorpromazine (CPZ) treatment or knockdown of the clathrin heavy chain (CLTC) protein significantly inhibited SVV infection. In addition, overexpression of CLTC promoted SVV infection. Caveolin-1 and membrane cholesterol were also required for SVV endocytosis. Notably, utilizing genistein, EIPA or nocodazole, we observed that macropinocytosis and microtubules are not involved in SVV entry. Furthermore, overexpression of the Rab7 and Rab9 proteins but not the Rab5 or Rab11 proteins promoted SVV infection. The findings were further validated by the knockdown of four Rabs and Lamp1 proteins, indicating that after internalization, SVV is transported from late endosomes to the trans-Golgi network (TGN) or lysosomes, respectively, eventually releasing its RNA into the cytosol from the lysosomes. Our findings concretely revealed SVV endocytosis mechanisms in HEK293T cells and provided an insightful theoretical foundation for further research into SVV oncolytic mechanisms.
Collapse
Affiliation(s)
- Wenqiang Liu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Xianfei Shang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Wei Wen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Xujiao Ren
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Liuxing Qin
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Xiangmin Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, PR China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, Hubei, PR China
| | - Ping Qian
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, PR China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, PR China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, Hubei, PR China
| |
Collapse
|
49
|
Steffen A, Reusch B, Gruteser N, Mainz D, Roncarati R, Baumann A, Stradal TEB, Knebel-Mörsdorf D. Baculovirus Actin Rearrangement-Inducing Factor 1 Can Remodel the Mammalian Actin Cytoskeleton. Microbiol Spectr 2023; 11:e0518922. [PMID: 36779726 PMCID: PMC10100760 DOI: 10.1128/spectrum.05189-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 01/23/2023] [Indexed: 02/14/2023] Open
Abstract
The actin rearrangement-inducing factor 1 (Arif-1) of Autographa californica multiple nucleopolyhedrovirus (AcMNPV) is an early viral protein that manipulates the actin cytoskeleton of host insect cells. Arif-1 is conserved among alphabaculoviruses and is responsible for the accumulation of F-actin at the plasma membrane during the early phase of infection. However, the molecular mechanism underlying Arif-1-induced cortical actin accumulation is still open. Recent studies have demonstrated the formation of invadosome-like structures induced by Arif-1, suggesting a function in systemic virus spread. Here, we addressed whether Arif-1 is able to manipulate the actin cytoskeleton of mammalian cells comparably to insect cells. Strikingly, transient overexpression of Arif-1 in B16-F1 mouse melanoma cells revealed pronounced F-actin remodeling. Actin assembly was increased, and intense membrane ruffling occurred at the expense of substrate-associated lamellipodia. Deletion mutagenesis studies of Arif-1 confirmed that the C-terminal cytoplasmic region was not sufficient to induce F-actin remodeling, supporting that the transmembrane region for Arif-1 function is also required in mammalian cells. The similarities between Arif-1-induced actin remodeling in insect and mammalian cells indicate that Arif-1 function relies on conserved cellular interaction partners and signal transduction pathways, thus providing an experimental tool to elucidate the underlying mechanism. IMPORTANCE Virus-induced changes of the host cell cytoskeleton play a pivotal role in the pathogenesis of viral infections. The baculovirus Autographa californica multiple nucleopolyhedrovirus (AcMNPV) is known for intervening with the regulation of the host actin cytoskeleton in a wide manner throughout the infection cycle. The actin rearrangement-inducing factor 1 (Arif-1) is a viral protein that causes actin rearrangement during the early phase of AcMNPV infection. Here, we performed overexpression studies of Arif-1 in mammalian cells to establish an experimental tool that allows elucidation of the mechanism underlying the Arif-1-induced remodeling of actin dynamics in a well-characterized and genetically accessible system.
Collapse
Affiliation(s)
- Anika Steffen
- Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Björn Reusch
- Center for Biochemistry, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nadine Gruteser
- Institute of Biological Information Processing, Molecular and Cellular Physiology, Research Center Juelich, Juelich, Germany
| | - Daniela Mainz
- Center for Biochemistry, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Renza Roncarati
- Center for Biochemistry, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Arnd Baumann
- Institute of Biological Information Processing, Molecular and Cellular Physiology, Research Center Juelich, Juelich, Germany
| | - Theresia E. B. Stradal
- Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dagmar Knebel-Mörsdorf
- Center for Biochemistry, University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Pediatrics, University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
50
|
TRIM40 is a pathogenic driver of inflammatory bowel disease subverting intestinal barrier integrity. Nat Commun 2023; 14:700. [PMID: 36755029 PMCID: PMC9908899 DOI: 10.1038/s41467-023-36424-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
The cortical actin cytoskeleton plays a critical role in maintaining intestinal epithelial integrity, and the loss of this architecture leads to chronic inflammation, as seen in inflammatory bowel disease (IBD). However, the exact mechanisms underlying aberrant actin remodeling in pathological states remain largely unknown. Here, we show that a subset of patients with IBD exhibits substantially higher levels of tripartite motif-containing protein 40 (TRIM40), a gene that is hardly detectable in healthy individuals. TRIM40 is an E3 ligase that directly targets Rho-associated coiled-coil-containing protein kinase 1 (ROCK1), an essential kinase involved in promoting cell-cell junctions, markedly decreasing the phosphorylation of key signaling factors critical for cortical actin formation and stabilization. This causes failure of the epithelial barrier function, thereby promoting a long-lived inflammatory response. A mutant TRIM40 lacking the RING, B-box, or C-terminal domains has impaired ability to accelerate ROCK1 degradation-driven cortical actin disruption. Accordingly, Trim40-deficient male mice are highly resistant to dextran sulfate sodium (DSS)-induced colitis. Our findings highlight that aberrant upregulation of TRIM40, which is epigenetically silenced under healthy conditions, drives IBD by subverting cortical actin formation and exacerbating epithelial barrier dysfunction.
Collapse
|