1
|
Haase JA, Marzi A. Molecular virulence determinants of human-pathogenic filoviruses. Adv Virus Res 2025; 121:1-29. [PMID: 40379380 DOI: 10.1016/bs.aivir.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2025]
Abstract
The Filoviridae family encompasses Ebola virus (EBOV) and Marburg virus (MARV), some of the most lethal viruses known to cause sporadic, recurring outbreaks of severe hemorrhagic fever mainly throughout central Africa. However, other lesser-known viruses also belong to the filovirus family as they are closely related, such as Bundibugyo, Reston and Taï Forest virus. These viruses differ in their virulence in humans significantly: while EBOV and MARV show lethality in humans of up to 90 %, Reston virus appears to be avirulent in humans. Here, underlying molecular factors leading to differences in virulence via changes in filovirus entry, replication and immune evasion strategies are summarized and assessed. While the filovirus glycoprotein contributes towards virulence by facilitating entry into a wide variety of tissues, differences in virus-host interactions and replication efficacies lead to measurable variances of progeny virus production. Additionally, immune evasion strategies lead to alterations in replication efficacy thus changing who has the upper hand between the virus and the host. Understanding and unraveling the contributions of these molecular determinants on filovirus virulence provide insights into the processes causing the underlying pathogenesis. It will further help to assess the pathogenicity of newly discovered filoviruses. Finally, these molecular determinants and processes present attractive targets for therapeutic intervention and development of novel antiviral countermeasures.
Collapse
Affiliation(s)
- Jil A Haase
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States.
| |
Collapse
|
2
|
Neupane R, Malla S, Karthikeyan C, Asbhy CR, Boddu SHS, Jayachandra Babu R, Tiwari AK. Endocytic highways: Navigating macropinocytosis and other endocytic routes for precision drug delivery. Int J Pharm 2025; 673:125356. [PMID: 39956408 DOI: 10.1016/j.ijpharm.2025.125356] [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: 09/15/2024] [Revised: 12/22/2024] [Accepted: 02/11/2025] [Indexed: 02/18/2025]
Abstract
Drug molecules can reach intracellular targets by different mechanisms, such as passive diffusion, active transport, and endocytosis. Endocytosis is the process by which cells engulf extracellular material by forming a vesicle and transporting it into the cells. In addition to its biological functions, endocytosis plays a vital role in the internalization of the therapeutic molecules. Clathrin-mediated endocytosis, caveolar endocytosis, and macropinocytosis are the most researched routes in the field of drug delivery. In addition to conventional small therapeutic molecules, the use of nanoformulations and large molecules, such as nucleic acids, peptides, and antibodies, have broadened the field of drug delivery. Although the majority of small therapeutic molecules can enter cells via passive diffusion, large molecules, and advanced targeted delivery systems, such as nanoparticles, are internalized by the endocytic route. Therefore, it is imperative to understand the characteristics of the endocytic routes in greater detail to design therapeutic molecules or formulations for successful delivery to the intracellular targets. This review highlights the prospects and limitations of the major endocytic routes for drug delivery, with a major emphasis on macropinocytosis. Since macropinocytosis is a non-selective uptake of extracellular matrix, the selective induction of macropinocytosis, using compounds that induce macropinocytosis and modulate macropinosome trafficking pathways, could be a potential approach for the intracellular delivery of diverse therapeutic modalities. Furthermore, we have summarized the characteristics associated with the formulations or drug carriers that can affect the endocytic routes for cellular internalization. The techniques that are used to study the intracellular uptake processes of therapeutic molecules are briefly discussed. Finally, the major limitations for intracellular targeting, endo-lysosomal degradation, and different approaches that have been used in overcoming these limitations, are highlighted in this review.
Collapse
Affiliation(s)
- Rabin Neupane
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, OH 43614, USA
| | - Saloni Malla
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, OH 43614, USA
| | - Chandrabose Karthikeyan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak 484887, India
| | - Charles R Asbhy
- Department of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, St. John's University, Queens, NY 10049, USA
| | - Sai H S Boddu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - R Jayachandra Babu
- Department of Drug Discovery and Development, Auburn University, AL 36849, USA
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, OH 43614, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
3
|
Saadh MJ, Muhammad FA, Albadr RJ, Sanghvi G, Jyothi SR, Kundlas M, Joshi KK, Gulyamov S, Taher WM, Alwan M, Jawad MJ, Al-Nuaimi AMA. From protein to immunology: comprehensive insights into Marburg virus vaccines, mechanism, and application. Arch Microbiol 2025; 207:74. [PMID: 40025302 DOI: 10.1007/s00203-025-04277-4] [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: 01/02/2025] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 03/04/2025]
Abstract
The Marburg virus (MARV), a member of the Filoviridae family, is a highly lethal pathogen that causes Marburg virus disease (MVD), a severe hemorrhagic fever with high fatality rates.Despite recurrent outbreaks, no licensed vaccine is currently available. This review explores MARV's genomic architecture, structural proteins, and recent advancements in vaccine development. It highlights the crucial role of MARV's seven monocistronic genes in viral replication and pathogenesis, with a focus on structural proteins such as nucleoprotein (NP), glycoprotein (GP), and viral proteins VP35, VP40, and VP24. These proteins are essential for viral entry, immune evasion, and replication. The review further examines various vaccine platforms, including multi-epitope vaccines, DNA-based vaccines, viral vector vaccines, virus-like particles (VLPs), and mRNA vaccines. Cutting-edge immunoinformatics approaches are discussed for identifying conserved epitopes critical for broad-spectrum protection. The immunological responses induced by these vaccine candidates, particularly their efficacy in preclinical trials, are analyzed, showcasing promising results in generating both humoral and cellular immunity. Moreover, the review addresses challenges and future directions in MARV vaccine development, emphasizing the need for enhanced immunogenicity, safety, and global accessibility. The integration of omics technologies (genomics, transcriptomics, proteomics) with immunoinformatics is presented as a transformative approach for next-generation vaccine design. Innovative platforms such as mRNA and VLP-based vaccines offer rapid and effective development opportunities. In this study, underscores the urgent need for a licensed MARV vaccine to prevent future outbreaks and strengthen global preparedness. By synthesizing the latest research and technological advancements, it provides a strategic roadmap for developing safe, effective, and broadly protective vaccines. The fight against MARV is a global priority, requiring coordinated efforts from researchers, policymakers, and public health organizations.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat, 360003, India
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Mayank Kundlas
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, Uttarakhand, 248002, India
- Graphic Era Deemed to Be University, Dehradun, Uttarakhand, India
| | - Surat Gulyamov
- Department of Dentistry and Pediatric Dentistry, Tashkent Pediatric Medical Institute, Bogishamol Street 223, 100140, Tashkent, Uzbekistan
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | |
Collapse
|
4
|
Cadena-Cruz C, De-Avila-Arias M, Costello HM, Hurtado-Gomez L, Martínez-De-La-Rosa W, Macchia-Ceballos G, Rosales-Rada W, Valencia-Villa G, Villalba-Amarís P, Kararoudi MN, Peeples ME, San-Juan-Vergara H. Respiratory syncytial virus fuses with plasma membrane to infect primary cultures of bronchial epithelial cells. Front Microbiol 2025; 16:1498955. [PMID: 40099186 PMCID: PMC11911548 DOI: 10.3389/fmicb.2025.1498955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/27/2025] [Indexed: 03/19/2025] Open
Abstract
Background Respiratory syncytial virus (RSV) is a common cause of bronchiolitis in children under the age of five. RSV infection proceeds by fusion of the viral envelope with the target cell membrane, but it is unclear whether fusion occurs with plasma or endosomal membranes. Methods Entry and/or infection was studied in undifferentiated primary cultures of human bronchial epithelial cells. Synchronization of viral entry or infection was achieved by attaching the virus to the plasma membrane at temperatures of 4°C or 22°C. Cells in which entry events had occurred were identified by the enzymatic action of beta-lactamase M (BlaM) fused to the RSV P protein (BlaM-P) carried by rgRSV virions. BlaM cleaves the beta-lactam ring of CCF2 loaded into the cells, disrupting FRET and allowing blue light to be emitted. Green fluorescent protein (GFP) expression, encoded by the rgRSV genome, was used to identify infected cells. Results We found that adsorption of RSV at 4°C favors entry via endocytosis, whereas binding of the virus to the membrane at 22°C favors RSV entry via the plasma membrane. The induction of endocytosis by synchronization at 4°C is, therefore, an artifact. In addition, we found that all drugs that interfered with RSV infection reduced cell membrane deformations such as filopodia and lamellipodia, suggesting a mechanism by which they may interfere with RSV fusion with the cell membrane. Discussion In conclusion, RSV enters the cell by direct fusion of its envelope with the plasma membrane.
Collapse
Affiliation(s)
- Christian Cadena-Cruz
- Departamento de Medicina, Universidad del Norte, Barranquilla, Colombia
- Programa de Bacteriología, Universidad Libre Seccional, Barranquilla, Colombia
| | | | - Heather M Costello
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | | | | | | | - Wendy Rosales-Rada
- Departamento de Medicina, Universidad del Norte, Barranquilla, Colombia
- Grupo de Investigación Avanzada en Biomedicina, Programa de Microbiología, Universidad Libre de Colombia, Barranquilla, Atlántico, Colombia
| | | | | | - Meisam Naeimi Kararoudi
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Mark E Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States
- Infectious Disease Institute, The Ohio State University, Columbus, OH, United States
| | | |
Collapse
|
5
|
Liang QZ, Ji CM, Wang B, Chen W, Cong F, Huang Y, Huang YW. Deltacoronavirus HKU11, HKU13, PDCoV (HKU15) and HKU17 spike pseudoviruses enter avian DF-1 cells via clathrin-mediated endocytosis in a Rab5-, Rab7- and pH-dependent manner. Vet Res 2025; 56:15. [PMID: 39825424 PMCID: PMC11740469 DOI: 10.1186/s13567-024-01442-3] [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: 08/21/2024] [Accepted: 11/14/2024] [Indexed: 01/20/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV), also known as HKU15, is a swine enteropathogenic virus that is believed to have originated in birds. PDCoV belongs to the genus Deltacoronavirus (DCoV), the members of which have mostly been identified in diverse avian species. We recently reported that chicken or porcine aminopeptidase N (APN), the major cellular receptor for PDCoV, can mediate cellular entry via three pseudotyped retroviruses displaying spike proteins from three avian DCoVs (HKU11, HKU13, and HKU17). In the present work, to better understand how avian-origin CoVs may be transmitted to pigs, we investigated the unknown DCoV entry pathway in avian cells. We show that clathrin-mediated endocytosis is involved in the entry of these DCoV pseudoviruses into chicken-origin DF-1 cells. Pseudovirus entry was suppressed by means of pharmacological inhibitors, dominant-negative mutants, and siRNAs targeting various cellular proteins and signalling molecules, suggesting that PDCoV and avian DCoV pseudovirus entry into DF-1 cells depends on clathrin, dynamin-2, cathepsins and a low-pH environment but is independent of caveolae and macropinocytosis. Furthermore, we found that DCoV pseudovirus entry was linked to Rab5- and Rab7-dependent pathways. This is the first report demonstrating that these DCoVs utilize clathrin-mediated endocytosis pathways to enter avian-origin cells, providing new insights into interspecies transmission of DCoVs.
Collapse
Affiliation(s)
- Qi-Zhang Liang
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, China
| | - Chun-Miao Ji
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| | - Bin Wang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Wei Chen
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, China
| | - Feng Cong
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China.
| | - Yu Huang
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, China.
| | - Yao-Wei Huang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China.
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
6
|
Wang LL, Seravalli J, Eaton B, Liu Y, Holbrook MR, Lan WJ, Xiang SH. Identification of Filovirus Entry Inhibitors from Marine Fungus-Derived Indole Alkaloids. Mar Drugs 2025; 23:23. [PMID: 39852525 PMCID: PMC11766795 DOI: 10.3390/md23010023] [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: 12/04/2024] [Revised: 12/28/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Filoviruses, mainly consisting of the two genera of Ebolavirus and Marburgvirus, are enveloped negative-strand RNA viruses that can infect humans to cause severe hemorrhagic fevers and outbreaks with high mortality rates. However, we still do not have effective medicines for treating these diseases. To search for effective drugs, we have identified three marine indole alkaloids that exhibit potent activities against filovirus infection. Thus, it is suggested that marine indole alkaloids can be a valuable compound source for filovirus drug screening and development. Since marine indole alkaloids comprise a large diverse group of secondary metabolites, their biological properties would be helpful for pharmaceutical drug development to treat various filovirus infections.
Collapse
Affiliation(s)
- Leah Liu Wang
- Nebraska Center for Virology, School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Javier Seravalli
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Brett Eaton
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, Frederick, MD 21702, USA
| | - Yi Liu
- Holland Computing Center, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Michael R. Holbrook
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, Frederick, MD 21702, USA
| | - Wen-Jian Lan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shi-Hua Xiang
- Nebraska Center for Virology, School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| |
Collapse
|
7
|
Nanbo A, Sakaguchi M, Furuyama W. Visualizing the Internalization of Marburg Viruslike Particles into Living Cells. Methods Mol Biol 2025; 2877:75-90. [PMID: 39585615 DOI: 10.1007/978-1-0716-4256-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Viral entry into cells is a pivotal stage of the infection process and, therefore, a prime target for the development of antiviral therapeutics. Here, we describe a system to monitor the internalization of lipophilic dye-labeled Marburg viruslike particles (VLPs) into living cells. Using cells stably expressing fluorescent protein-fused markers for specific cell organelles, the VLP entry process can be visualized. This procedure enables the characterization of the entry process by visualizing individual steps using specific bio-probes. Additionally, when combined with image analysis, this method allows for the quantification of the efficiencies of individual entry steps including particle adsorption, uptake by endocytosis, and membrane fusion. Finally, this method can be used for antiviral drug screening.
Collapse
Affiliation(s)
- Asuka Nanbo
- National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki, Japan.
| | - Miako Sakaguchi
- Central Laboratory, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Wakako Furuyama
- National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
8
|
Li W, Yang W, Liu X, Zhou W, Wang S, Wang Z, Zhao Y, Feng N, Wang T, Wu M, Ge L, Xia X, Yan F. Fully human monoclonal antibodies against Ebola virus possess complete protection in a hamster model. Emerg Microbes Infect 2024; 13:2392651. [PMID: 39155772 PMCID: PMC11348817 DOI: 10.1080/22221751.2024.2392651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/14/2024] [Accepted: 08/11/2024] [Indexed: 08/20/2024]
Abstract
Ebola disease is a lethal viral hemorrhagic fever caused by ebolaviruses within the Filoviridae family with mortality rates of up to 90%. Monoclonal antibody (mAb) based therapies have shown great potential for the treatment of EVD. However, the potential emerging ebolavirus isolates and the negative effect of decoy protein on the therapeutic efficacy of antibodies highlight the necessity of developing novel antibodies to counter the threat of Ebola. Here, 11 fully human mAbs were isolated from transgenic mice immunized with GP protein and recombinant vesicular stomatitis virus-bearing GP (rVSV-EBOV GP). These mAbs were divided into five groups according to their germline genes and exhibited differential binding activities and neutralization capabilities. In particular, mAbs 8G6, 2A4, and 5H4 were cross-reactive and bound at least three ebolavirus glycoproteins. mAb 4C1 not only exhibited neutralizing activity but no cross-reaction with sGP. mAb 7D8 exhibited the strongest neutralizing capacity. Further analysis on the critical residues for the bindings of 4C1 and 8G6 to GPs was conducted using antibodies complementarity-determining regions (CDRs) alanine scanning. It has been shown that light chain CDR3 played a crucial role in binding and neutralization and that any mutation in CDRs could not improve the binding of 4C1 to sGP. Importantly, mAbs 7D8, 8G6, and 4C1 provided complete protections against EBOV infection in a hamster lethal challenge model when administered 12 h post-infection. These results support mAbs 7D8, 8G6, and 4C1 as potent antibody candidates for further investigations and pave the way for further developments of therapies and vaccines.
Collapse
Affiliation(s)
- Wujian Li
- College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Wanying Yang
- Department of Laboratory Animal Science, Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Xueqin Liu
- Chongqing Academy of Animal Sciences, Chongqing, People’s Republic of China
| | - Wujie Zhou
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Shen Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Zhenshan Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Yongkun Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Na Feng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Tiecheng Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Meng Wu
- Chongqing Academy of Animal Sciences, Chongqing, People’s Republic of China
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences, Chongqing, People’s Republic of China
| | - Xianzhu Xia
- College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| | - Feihu Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| |
Collapse
|
9
|
Wang J, Ziarnik M, Zhang XF, Jagota A. Biomechanics Model to Characterize Atomic Force Microscopy-Based Virus-Host Cell Adhesion Measurements. J Phys Chem B 2024; 128:11546-11553. [PMID: 39316705 PMCID: PMC11613445 DOI: 10.1021/acs.jpcb.4c04527] [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: 07/06/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024]
Abstract
We present a model for virus-cell adhesion that can be used for quantitative extraction of adhesive properties from atomic force microscopy (AFM)-based force spectroscopy measurements. We extend a previously reported continuum model of viral cell interactions based on a single parameter representing adhesive energy density by using a cohesive zone model in which adhesion is represented by two parameters, a pull-off stress and associated characteristic displacement. This approach accounts for the deformability of the adhesive receptors, such as the Spike protein and transmembrane immunoglobulin and mucin domain (TIM) family that mediate adhesion of SARS-CoV-2 and Ebola viruses, and the omnipresent glycocalyx. Our model represents receptors as a Winkler foundation and aims to predict the pull-off force needed to break the adhesion between the virus and the cell. By comparing the force-separation curves simulated by the model and experimental data, we found that the model can effectively explain the AFM pull-off force trace, thus allowing quantification of the adhesion parameters. Our model provides a more refined understanding of viral cell adhesion and also establishes a framework for interpreting and predicting AFM force spectroscopy measurements.
Collapse
Affiliation(s)
- Jiajun Wang
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Matthew Ziarnik
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - X. Frank Zhang
- Department
of Biomedical Engineering, University of
Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Anand Jagota
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
- Department
of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
10
|
Wang S, Cheng P, Guo K, Ren S, Tadele BA, Liang Z, Sun Y, Yin X, Wang X. Lumpy skin disease virus enters into host cells via dynamin-mediated endocytosis and macropinocytosis. Vet Microbiol 2024; 298:110254. [PMID: 39307114 DOI: 10.1016/j.vetmic.2024.110254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 11/19/2024]
Abstract
Lumpy skin disease virus (LSDV), a ruminant poxvirus of the Capripoxvirus genus, is the etiologic agent of an economically important cattle disease categorized as a notifiable disease by the World Organization for Animal Health. However, the endocytic pathway and their regulatory molecules have not been characterized for LSDV. In the present study, specific pharmacological inhibitors were used to analyze the mechanism of LSDV entry into Mardin-Darby Bovine Kidney cell (MDBK) and bovine mammary epithelial cell (BMEC). The results showed that LSDV entered MDBK and BMEC cells depends on low-pH conditions and dynamin. However, the inhibitor of caveolae- and clathrin-mediated endocytosis cann't inhibit LSDV entry into MDBK and BMEC cells. Furthermore, treatment with specific inhibitors demonstrated that LSDV entry into MDBK and BMEC cells via macropinocytosis depended on the Na1/H1 exchanger (NHE) but not phosphatidylinositol 3-kinase (PI3K). In addition, results demonstrated that these inhibitors inhibited LSDV entry but did not have effect on LSDV binding. Taken together, our study demonstrated that LSDV enters MDBK and BMEC cells through macropinocytosis pathway in a low-PH- and dynamin-dependent manner while independent on PI3K. Results presented in this study potentially provides insight into the entry mechanisms of LSDV, and it may facilitate the development of therapeutic interventions.
Collapse
Affiliation(s)
- Shasha Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Pengyuan Cheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Ke Guo
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Shanhui Ren
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Berihun Afera Tadele
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Mekelle University College of Veterinary Sciences, Mekelle, Tigray, Ethiopia
| | - Zhengji Liang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yuefeng Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiangping Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiangwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.
| |
Collapse
|
11
|
Hou D, Mu Q, Chen W, Cao W, Zhang XF. Nano-Biomechanical Investigation of Phosphatidylserine-Mediated Ebola Viral Attachment via Human Gas6 and Axl. Viruses 2024; 16:1700. [PMID: 39599815 PMCID: PMC11599018 DOI: 10.3390/v16111700] [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: 08/15/2024] [Revised: 10/20/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
The Ebola virus is a deadly pathogen that has been threatening public health for decades. Recent studies have revealed alternative viral invasion routes where Ebola virus approaches cells via interactions among phosphatidylserine (PS), PS binding ligands such as Gas6, and TAM family receptors such as Axl. In this study, we investigate the interactions among phosphatidylserine on the Ebola viral-like particle (VLP) membrane, human Gas6, and human Axl using atomic force microscope-based single molecule force spectroscopy to compare their binding strength and affinity from a biomechanical perspective. The impact of calcium ions on their interactions is also studied and quantified to provide more details on the calcium-dependent phosphatidylserine-Gas6 binding mechanism. Our results indicate that, in the presence of calcium ions, the binding strengths of VLP-Gas6 and VLP-Gas6-Axl increase but are still weaker than that of Gas6-Axl, and the binding affinity of VLP-Gas6 and VLP-Gas6-Axl is largely improved. The binding strength and affinity of Gas6-Axl basically remain the same, indicating no impact in the presence of calcium ions. Together, our study suggests that, under physiological conditions with calcium present, the Ebola virus can utilize its membrane phosphatidylserine to dock on cell surface via Gas6-Axl bound complex.
Collapse
Affiliation(s)
- Decheng Hou
- Department of Bioengineering, Lehigh University, Bethlehem, PA 18015, USA;
- Department of Biomedical Engineering, University of Massachusetts at Amherst, Amherst, MA 01003, USA; (Q.M.); (W.C.)
| | - Qian Mu
- Department of Biomedical Engineering, University of Massachusetts at Amherst, Amherst, MA 01003, USA; (Q.M.); (W.C.)
| | - Weixuan Chen
- Department of Biomedical Engineering, University of Massachusetts at Amherst, Amherst, MA 01003, USA; (Q.M.); (W.C.)
| | - Wenpeng Cao
- Department of Bioengineering, Lehigh University, Bethlehem, PA 18015, USA;
| | - Xiaohui Frank Zhang
- Department of Bioengineering, Lehigh University, Bethlehem, PA 18015, USA;
- Department of Biomedical Engineering, University of Massachusetts at Amherst, Amherst, MA 01003, USA; (Q.M.); (W.C.)
| |
Collapse
|
12
|
Wang S, Li W, Wang Z, Yang W, Li E, Xia X, Yan F, Chiu S. Emerging and reemerging infectious diseases: global trends and new strategies for their prevention and control. Signal Transduct Target Ther 2024; 9:223. [PMID: 39256346 PMCID: PMC11412324 DOI: 10.1038/s41392-024-01917-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/13/2024] [Accepted: 07/05/2024] [Indexed: 09/12/2024] Open
Abstract
To adequately prepare for potential hazards caused by emerging and reemerging infectious diseases, the WHO has issued a list of high-priority pathogens that are likely to cause future outbreaks and for which research and development (R&D) efforts are dedicated, known as paramount R&D blueprints. Within R&D efforts, the goal is to obtain effective prophylactic and therapeutic approaches, which depends on a comprehensive knowledge of the etiology, epidemiology, and pathogenesis of these diseases. In this process, the accessibility of animal models is a priority bottleneck because it plays a key role in bridging the gap between in-depth understanding and control efforts for infectious diseases. Here, we reviewed preclinical animal models for high priority disease in terms of their ability to simulate human infections, including both natural susceptibility models, artificially engineered models, and surrogate models. In addition, we have thoroughly reviewed the current landscape of vaccines, antibodies, and small molecule drugs, particularly hopeful candidates in the advanced stages of these infectious diseases. More importantly, focusing on global trends and novel technologies, several aspects of the prevention and control of infectious disease were discussed in detail, including but not limited to gaps in currently available animal models and medical responses, better immune correlates of protection established in animal models and humans, further understanding of disease mechanisms, and the role of artificial intelligence in guiding or supplementing the development of animal models, vaccines, and drugs. Overall, this review described pioneering approaches and sophisticated techniques involved in the study of the epidemiology, pathogenesis, prevention, and clinical theatment of WHO high-priority pathogens and proposed potential directions. Technological advances in these aspects would consolidate the line of defense, thus ensuring a timely response to WHO high priority pathogens.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Wujian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Zhenshan Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Wanying Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China.
- Department of Laboratory Medicine, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
13
|
Furuyama W, Yamada K, Sakaguchi M, Marzi A, Nanbo A. Marburg virus exploits the Rab11-mediated endocytic pathway in viral-particle production. Microbiol Spectr 2024; 12:e0026924. [PMID: 39078193 PMCID: PMC11370620 DOI: 10.1128/spectrum.00269-24] [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: 03/12/2024] [Accepted: 06/10/2024] [Indexed: 07/31/2024] Open
Abstract
Filoviruses produce viral particles with characteristic filamentous morphology. The major viral matrix protein, VP40, is trafficked to the plasma membrane and promotes viral particle formation and subsequent viral egress. In the present study, we assessed the role of the small GTPase Rab11-mediated endocytic pathway in Marburg virus (MARV) particle formation and budding. Although Rab11 was predominantly localized in the perinuclear region, it exhibited a more diffuse distribution in the cytoplasm of cells transiently expressing MARV VP40. Rab11 was incorporated into MARV-like particles. Expression of the dominant-negative form of Rab11 and knockdown of Rab11 decreased the amount of VP40 fractions in the cell periphery. Moreover, downregulation of Rab11 moderately reduced the release of MARV-like particles and authentic MARV. We further demonstrated that VP40 induces the distribution of the microtubule network toward the cell periphery, which was partly associated with Rab11. Depolymerization of microtubules reduced the accumulation of VP40 in the cell periphery along with viral particle formation. VP40 physically interacted with α-tubulin, a major component of microtubules, but not with Rab11. Taken together, these results suggested that VP40 partly interacts with microtubules and facilitates their distribution toward the cell periphery, leading to the trafficking of transiently tethering Rab11-positive vesicles toward the cell surface. As we previously demonstrated the role of Rab11 in the formation of Ebola virus particles, the results here suggest that filoviruses in general exploit the vesicle-trafficking machinery for proper virus-particle formation and subsequent egress. These pathways may be a potential target for the development of pan-filovirus therapeutics.IMPORTANCEFiloviruses, including Marburg and Ebola viruses, produce distinct filamentous viral particles. Although it is well known that the major viral matrix protein of these viruses, VP40, is trafficked to the cell surface and promotes viral particle production, details regarding the associated molecular mechanisms remain unclear. To address this knowledge gap, we investigated the role of the small GTPase Rab11-mediated endocytic pathway in this process. Our findings revealed that Marburg virus exploits the Rab11-mediated vesicle-trafficking pathway for the release of virus-like particles and authentic virions in a microtubule network-dependent manner. Previous findings demonstrated that Rab11 is also involved in Ebola virus-particle production. Taken together, these data suggest that filoviruses, in general, may hijack the microtubule-dependent vesicle-trafficking machinery for productive replication. Therefore, this pathway presents as a potential target for the development of pan-filovirus therapeutics.
Collapse
Affiliation(s)
- Wakako Furuyama
- National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki, Japan
| | - Kento Yamada
- National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki, Japan
| | - Miako Sakaguchi
- Central Laboratory, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Asuka Nanbo
- National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
14
|
Li Y, Wang X, Bi Y, Zhang M, Xiong W, Hu X, Zhang Y, He F. SNX5-Rab11a protects against cardiac hypertrophy through regulating LRP6 membrane translocation. J Mol Cell Cardiol 2024; 194:46-58. [PMID: 38950816 DOI: 10.1016/j.yjmcc.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 06/20/2024] [Accepted: 06/28/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUNDS Pathological cardiac hypertrophy is considered one of the independent risk factors for heart failure, with a rather complex pathogenic machinery. Sorting nexins (SNXs), denoting a diverse family of cytoplasmic- and membrane-associated phosphoinositide-binding proteins, act as a pharmacological target against specific cardiovascular diseases including heart failure. Family member SNX5 was reported to play a pivotal role in a variety of biological processes. However, contribution of SNX5 to the development of cardiac hypertrophy, remains unclear. METHODS Mice underwent transverse aortic constriction (TAC) to induce cardiac hypertrophy and simulate pathological conditions. TAC model was validated using echocardiography and histological staining. Expression of SNX5 was assessed by western blotting. Then, SNX5 was delivered through intravenous administration of an adeno-associated virus serotype 9 carrying cTnT promoter (AAV9-cTnT-SNX5) to achieve SNX5 cardiac-specific overexpression. To assess the impact of SNX5, morphological analysis, echocardiography, histological staining, hypertrophic biomarkers, and cardiomyocyte contraction were evaluated. To unravel potential molecular events associated with SNX5, interactome analysis, fluorescence co-localization, and membrane protein profile were evaluated. RESULTS Our results revealed significant downregulated protein level of SNX5 in TAC-induced hypertrophic hearts in mice. Interestingly, cardiac-specific overexpression of SNX5 improved cardiac function, with enhanced left ventricular ejection fraction, fraction shortening, as well as reduced cardiac fibrosis. Mechanistically, SNX5 directly bound to Rab11a, increasing membrane accumulation of Rab11a (a Rab GTPase). Afterwards, this intricate molecular interaction upregulated the membrane content of low-density lipoprotein receptor-related protein 6 (LRP6), a key regulator against cardiac hypertrophy. Our comprehensive assessment of siRab11a expression in HL-1 cells revealed its role in antagonism of LRP6 membrane accumulation under SNX5 overexpression. CONCLUSIONS This study revealed that binding of SNX5 with LRP6 triggers their membrane translocation through Rab11a assisting, defending against cardiac remodeling and cardiac dysfunction under pressure overload. These findings provide new insights into the previously unrecognized role of SNX5 in the progression of cardiac hypertrophy.
Collapse
Affiliation(s)
- Yutong Li
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, Anhui 230601, China; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Xiang Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yaguang Bi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Mengjiao Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Weidong Xiong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Xiaolong Hu
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, Anhui 230601, China
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| | - Fei He
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, Anhui 230601, China.
| |
Collapse
|
15
|
Liang J, Djurkovic MA, Leavitt CG, Shtanko O, Harty RN. Hippo signaling pathway regulates Ebola virus transcription and egress. Nat Commun 2024; 15:6953. [PMID: 39138205 PMCID: PMC11322314 DOI: 10.1038/s41467-024-51356-z] [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: 01/24/2024] [Accepted: 08/02/2024] [Indexed: 08/15/2024] Open
Abstract
Filovirus-host interactions play important roles in all stages of the virus lifecycle. Here, we identify LATS1/2 kinases and YAP, key components of the Hippo pathway, as critical regulators of EBOV transcription and egress. Specifically, we find that when YAP is phosphorylated by LATS1/2, it localizes to the cytoplasm (Hippo "ON") where it sequesters VP40 to prevent egress. In contrast, when the Hippo pathway is "OFF", unphosphorylated YAP translocates to the nucleus where it transcriptionally activates host genes and promotes viral egress. Our data reveal that LATS2 indirectly modulates filoviral VP40-mediated egress through phosphorylation of AMOTp130, a positive regulator of viral egress, but more surprisingly that LATS1/2 kinases directly modulate EBOV transcription by phosphorylating VP30, an essential regulator of viral transcription. In sum, our findings highlight the potential to exploit the Hippo pathway/filovirus axis for the development of host-oriented countermeasures targeting EBOV and related filoviruses.
Collapse
Affiliation(s)
- Jingjing Liang
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA, 19104, USA
| | - Marija A Djurkovic
- Host-Pathogen Interactions, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX, 78227, USA
| | - Carson G Leavitt
- Host-Pathogen Interactions, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX, 78227, USA
| | - Olena Shtanko
- Host-Pathogen Interactions, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX, 78227, USA.
| | - Ronald N Harty
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA, 19104, USA.
| |
Collapse
|
16
|
Gong M, Peng C, Yang C, Wang Z, Qian H, Hu X, Zhou P, Shan C, Ding Q. Genome-wide CRISPR/Cas9 screen identifies SLC39A9 and PIK3C3 as crucial entry factors for Ebola virus infection. PLoS Pathog 2024; 20:e1012444. [PMID: 39173055 PMCID: PMC11341029 DOI: 10.1371/journal.ppat.1012444] [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: 02/17/2024] [Accepted: 07/23/2024] [Indexed: 08/24/2024] Open
Abstract
The Ebola virus (EBOV) has emerged as a significant global health concern, notably during the 2013-2016 outbreak in West Africa. Despite the clinical approval of two EBOV antibody drugs, there is an urgent need for more diverse and effective antiviral drugs, along with comprehensive understanding of viral-host interactions. In this study, we harnessed a biologically contained EBOVΔVP30-EGFP cell culture model which could recapitulate the entire viral life cycle, to conduct a genome-wide CRISPR/Cas9 screen. Through this, we identified PIK3C3 (phosphatidylinositide 3-kinase) and SLC39A9 (zinc transporter) as crucial host factors for EBOV infection. Genetic depletion of SLC39A9 and PIK3C3 lead to reduction of EBOV entry, but not impact viral genome replication, suggesting that SLC39A9 and PIK3C3 act as entry factors, facilitating viral entry into host cells. Moreover, PIK3C3 kinase activity is indispensable for the internalization of EBOV virions, presumably through the regulation of endocytic and autophagic membrane traffic, which has been previously recognized as essential for EBOV internalization. Notably, our study demonstrated that PIK3C3 kinase inhibitor could effectively block EBOV infection, underscoring PIK3C3 as a promising drug target. Furthermore, biochemical analysis showed that recombinant SLC39A9 protein could directly bind viral GP protein, which further promotes the interaction of viral GP protein with cellular receptor NPC1. These findings suggests that SLC39A9 plays dual roles in EBOV entry. Initially, it serves as an attachment factor during the early entry phase by engaging with the viral GP protein. Subsequently, SLC39A9 functions an adaptor protein, facilitating the interaction between virions and the NPC1 receptor during the late entry phase, prior to cathepsin cleavage on the viral GP. In summary, this study offers novel insights into virus-host interactions, contributing valuable information for the development of new therapies against EBOV infection.
Collapse
Affiliation(s)
- Mingli Gong
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Cheng Peng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Chen Yang
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Zhenhua Wang
- The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongwu Qian
- The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Peng Zhou
- CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Chao Shan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- CAS Key Laboratory of Special Pathogens, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Qiang Ding
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
17
|
Bagdonaite I, Abdurahman S, Mirandola M, Pasqual D, Frank M, Narimatsu Y, Joshi HJ, Vakhrushev SY, Salata C, Mirazimi A, Wandall HH. Targeting host O-linked glycan biosynthesis affects Ebola virus replication efficiency and reveals differential GalNAc-T acceptor site preferences on the Ebola virus glycoprotein. J Virol 2024; 98:e0052424. [PMID: 38757972 PMCID: PMC11237518 DOI: 10.1128/jvi.00524-24] [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: 03/21/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
Ebola virus glycoprotein (EBOV GP) is one of the most heavily O-glycosylated viral glycoproteins, yet we still lack a fundamental understanding of the structure of its large O-glycosylated mucin-like domain and to what degree the host O-glycosylation capacity influences EBOV replication. Using tandem mass spectrometry, we identified 47 O-glycosites on EBOV GP and found similar glycosylation signatures on virus-like particle- and cell lysate-derived GP. Furthermore, we performed quantitative differential O-glycoproteomics on proteins produced in wild-type HEK293 cells and cell lines ablated for the three key initiators of O-linked glycosylation, GalNAc-T1, -T2, and -T3. The data show that 12 out of the 47 O-glycosylated sites were regulated, predominantly by GalNAc-T1. Using the glycoengineered cell lines for authentic EBOV propagation, we demonstrate the importance of O-linked glycan initiation and elongation for the production of viral particles and the titers of progeny virus. The mapped O-glycan positions and structures allowed to generate molecular dynamics simulations probing the largely unknown spatial arrangements of the mucin-like domain. The data highlight targeting GALNT1 or C1GALT1C1 as a possible way to modulate O-glycan density on EBOV GP for novel vaccine designs and tailored intervention approaches.IMPORTANCEEbola virus glycoprotein acquires its extensive glycan shield in the host cell, where it is decorated with N-linked glycans and mucin-type O-linked glycans. The latter is initiated by a family of polypeptide GalNAc-transferases that have different preferences for optimal peptide substrates resulting in a spectrum of both very selective and redundant substrates for each isoform. In this work, we map the exact locations of O-glycans on Ebola virus glycoprotein and identify subsets of sites preferentially initiated by one of the three key isoforms of GalNAc-Ts, demonstrating that each enzyme contributes to the glycan shield integrity. We further show that altering host O-glycosylation capacity has detrimental effects on Ebola virus replication, with both isoform-specific initiation and elongation playing a role. The combined structural and functional data highlight glycoengineered cell lines as useful tools for investigating molecular mechanisms imposed by specific glycans and for steering the immune responses in future vaccine designs.
Collapse
Affiliation(s)
- Ieva Bagdonaite
- Department of Cellular and Molecular Medicine, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark
| | | | - Mattia Mirandola
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Denis Pasqual
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | - Yoshiki Narimatsu
- Department of Cellular and Molecular Medicine, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark
| | - Hiren J Joshi
- Department of Cellular and Molecular Medicine, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark
| | - Sergey Y Vakhrushev
- Department of Cellular and Molecular Medicine, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark
| | - Cristiano Salata
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Ali Mirazimi
- Public Health Agency of Sweden, Solna, Sweden
- Department of Laboratory Medicine (LABMED), Karolinska Institute, Stockholm, Sweden
- National Veterinary Institute, Uppsala, Sweden
| | - Hans H Wandall
- Department of Cellular and Molecular Medicine, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
18
|
Vaknin A, Grossman A, Durham ND, Lupovitz I, Goren S, Golani G, Roichman Y, Munro JB, Sorkin R. Ebola Virus Glycoprotein Strongly Binds to Membranes in the Absence of Receptor Engagement. ACS Infect Dis 2024; 10:1590-1601. [PMID: 38684073 PMCID: PMC11091876 DOI: 10.1021/acsinfecdis.3c00622] [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: 11/15/2023] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 05/02/2024]
Abstract
Ebola virus (EBOV) is an enveloped virus that must fuse with the host cell membrane in order to release its genome and initiate infection. This process requires the action of the EBOV envelope glycoprotein (GP), encoded by the virus, which resides in the viral envelope and consists of a receptor binding subunit, GP1, and a membrane fusion subunit, GP2. Despite extensive research, a mechanistic understanding of the viral fusion process is incomplete. To investigate GP-membrane association, a key step in the fusion process, we used two approaches: high-throughput measurements of single-particle diffusion and single-molecule measurements with optical tweezers. Using these methods, we show that the presence of the endosomal Niemann-Pick C1 (NPC1) receptor is not required for primed GP-membrane binding. In addition, we demonstrate this binding is very strong, likely attributed to the interaction between the GP fusion loop and the membrane's hydrophobic core. Our results also align with previously reported findings, emphasizing the significance of acidic pH in the protein-membrane interaction. Beyond Ebola virus research, our approach provides a powerful toolkit for studying other protein-membrane interactions, opening new avenues for a better understanding of protein-mediated membrane fusion events.
Collapse
Affiliation(s)
- Alisa Vaknin
- School
of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Center
for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Alon Grossman
- School
of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Center
for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Natasha D. Durham
- Department
of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, United States
| | - Inbal Lupovitz
- School
of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Center
for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shahar Goren
- School
of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Center
for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Gonen Golani
- Department
of Physics and Haifa Research Center for Theoretical Physics and Astrophysics, University of Haifa, Haifa 3498838, Israel
| | - Yael Roichman
- School
of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Center
for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv 6997801, Israel
- Raymond
and Beverly Sackler School of Physics & Astronomy, Tel Aviv University, Tel Aviv 6997801, Israel
| | - James B. Munro
- Department
of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, United States
- Department
of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605, United States
| | - Raya Sorkin
- School
of Chemistry, Raymond & Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Center
for Physics and Chemistry of Living Systems, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
19
|
Iyer K, Yan Z, Ross SR. Entry inhibitors as arenavirus antivirals. Front Microbiol 2024; 15:1382953. [PMID: 38650890 PMCID: PMC11033450 DOI: 10.3389/fmicb.2024.1382953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
Arenaviruses belonging to the Arenaviridae family, genus mammarenavirus, are enveloped, single-stranded RNA viruses primarily found in rodent species, that cause severe hemorrhagic fever in humans. With high mortality rates and limited treatment options, the search for effective antivirals is imperative. Current treatments, notably ribavirin and other nucleoside inhibitors, are only partially effective and have significant side effects. The high lethality and lack of treatment, coupled with the absence of vaccines for all but Junín virus, has led to the classification of these viruses as Category A pathogens by the Centers for Disease Control (CDC). This review focuses on entry inhibitors as potential therapeutics against mammarenaviruses, which include both New World and Old World arenaviruses. Various entry inhibition strategies, including small molecule inhibitors and neutralizing antibodies, have been explored through high throughput screening, genome-wide studies, and drug repurposing. Notable progress has been made in identifying molecules that target receptor binding, internalization, or fusion steps. Despite promising preclinical results, the translation of entry inhibitors to approved human therapeutics has faced challenges. Many have only been tested in in vitro or animal models, and a number of candidates showed efficacy only against specific arenaviruses, limiting their broader applicability. The widespread existence of arenaviruses in various rodent species and their potential for their zoonotic transmission also underscores the need for rapid development and deployment of successful pan-arenavirus therapeutics. The diverse pool of candidate molecules in the pipeline provides hope for the eventual discovery of a broadly effective arenavirus antiviral.
Collapse
Affiliation(s)
| | | | - Susan R. Ross
- Department of Microbiology and Immunology, University of Illinois, College of Medicine, Chicago, IL, United States
| |
Collapse
|
20
|
Soni M, Tulsian K, Barot P, Vyas VK. Recent Advances in Therapeutic Approaches Against Ebola Virus Infection. RECENT ADVANCES IN ANTI-INFECTIVE DRUG DISCOVERY 2024; 19:276-299. [PMID: 38279760 DOI: 10.2174/0127724344267452231206061944] [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: 06/15/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND Ebola virus (EBOV) is a genus of negative-strand RNA viruses belonging to the family Filoviradae that was first described in 1976 in the present-day Democratic Republic of the Congo. It has intermittently affected substantial human populations in West Africa and presents itself as a global health menace due to the high mortality rate of patients, high transmission rate, difficult patient management, and the emergence of complicated autoimmune disease-like conditions post-infection. OBJECTIVE EBOV or other EBOV-like species as a biochemical weapon pose a significant risk; hence, the need to develop both prophylactic and therapeutic medications to combat the virus is unquestionable. METHODS In this review work, we have compiled the literature pertaining to transmission, pathogenesis, immune response, and diagnosis of EBOV infection. We included detailed structural details of EBOV along with all the available therapeutics against EBOV disease. We have also highlighted current developments and recent advances in therapeutic approaches against Ebola virus disease (EVD). DISCUSSION The development of preventive vaccines against the virus is proving to be a successful effort as of now; however, problems concerning logistics, product stability, multi- dosing, and patient tracking are prominent in West Africa. Monoclonal antibodies that target EBOV proteins have also been developed and approved in the clinic; however, no small drug molecules that target these viral proteins have cleared clinical trials. An understanding of clinically approved vaccines and their shortcomings also serves an important purpose for researchers in vaccine design in choosing the right vector, antigen, and particular physicochemical properties that are critical for the vaccine's success against the virus across the world. CONCLUSION Our work brings together a comprehensive review of all available prophylactic and therapeutic medications developed and under development against the EBOV, which will serve as a guide for researchers in pursuing the most promising drug discovery strategies against the EBOV and also explore novel mechanisms of fighting against EBOV infection.
Collapse
Affiliation(s)
- Molisha Soni
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Kartik Tulsian
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Parv Barot
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Vivek Kumar Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| |
Collapse
|
21
|
Li S, Xiao D, Zhang L, Chen R, Song D, Wen Y, Wu R, Zhao Q, Du S, Yan Q, Cao S, Huang X. Porcine deltacoronavirus enters ST cells by clathrin-mediated endocytosis and does not require Rab5, Rab7, or Rab11. Microbiol Spectr 2023; 11:e0255323. [PMID: 37962380 PMCID: PMC10714841 DOI: 10.1128/spectrum.02553-23] [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: 06/20/2023] [Accepted: 09/29/2023] [Indexed: 11/15/2023] Open
Abstract
IMPORTANCE Porcine deltacoronavirus (PDCoV) is a newly emerged enteric virus threatening pig industries worldwide. Our previous work showed that PDCoV enters porcine kidney (PK-15) cells through a caveolae-dependent pathway, but the entry mechanism for PDCoV into swine testicle (ST) cells remains unclear. Mechanisms of virus entry can be different with different virus isolates and cell types. Here, we determined that PDCoV enters ST cells via clathrin-mediated endocytosis. Additionally, we found that PDCoV entry does not require Rab5, Rab7, or Rab11. These findings provide additional understanding of the entry mechanisms of PDCoV and possible antiviral targets.
Collapse
Affiliation(s)
- Shiqian Li
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Dai Xiao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Luwen Zhang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Rui Chen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Daili Song
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yiping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Rui Wu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qin Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Senyan Du
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qigui Yan
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Sanjie Cao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- National Animal Experiments Teaching Demonstration Center, Sichuan Agricultural University, Chengdu, China
| | - Xiaobo Huang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- National Animal Experiments Teaching Demonstration Center, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
22
|
Bi J, Wang H, Han Q, Pei H, Wang H, Jin H, Jin S, Chi H, Yang S, Zhao Y, Yan F, Ge L, Xia X. A rabies virus-vectored vaccine expressing two copies of the Marburg virus glycoprotein gene induced neutralizing antibodies against Marburg virus in humanized mice. Emerg Microbes Infect 2023; 12:2149351. [PMID: 36453198 PMCID: PMC9809360 DOI: 10.1080/22221751.2022.2149351] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Marburg virus disease (MVD) is a lethal viral haemorrhagic fever caused by Marburg virus (MARV) with a case fatality rate as high as 88%. There is currently no vaccine or antiviral therapy approved for MVD. Due to high variation among MARV isolates, vaccines developed against one strain fail to protect against other strains. Here we report that three recombinant rabies virus (RABV) vector vaccines encoding two copies of GPs covering both MARV lineages induced pseudovirus neutralizing antibodies in BALB/c mice. Furthermore, high-affinity human neutralizing antibodies were isolated from a humanized mouse model. The three vaccines produced a Th1-biased serological response similar to that of human patients. Adequate sequential immunization enhanced the production of neutralizing antibodies. Virtual docking suggested that neutralizing antibodies induced by the Angola strain seemed to be able to hydrogen bond to the receptor-binding site (RBS) in the GP of the Ravn strain through hypervariable regions 2 (CDR2) and CDR3 of the VH region. These findings demonstrate that three inactivated vaccines are promising candidates against different strains of MARV, and a novel fully humanized neutralizing antibody against MARV was isolated.
Collapse
Affiliation(s)
- Jinhao Bi
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Haojie Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Qiuxue Han
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China,Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, People’s Republic of China
| | - Hongyan Pei
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China,College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Hualei Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
| | - Hongli Jin
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
| | - Song Jin
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China,Ruminant Disease Research Center, College of Life Sciences, Shandong Normal University, Jinan, People’s Republic of China
| | - Hang Chi
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Songtao Yang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Yongkun Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Feihu Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China, Feihu Yan ; Liangpeng Ge ; Xianzhu Xia
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences, Chongqing, People’s Republic of China, Feihu Yan ; Liangpeng Ge ; Xianzhu Xia
| | - Xianzhu Xia
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China,Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, People’s Republic of China, Feihu Yan ; Liangpeng Ge ; Xianzhu Xia
| |
Collapse
|
23
|
Jain A, Govindan R, Berkman AR, Luban J, Díaz-Salinas MA, Durham ND, Munro JB. Regulation of Ebola GP conformation and membrane binding by the chemical environment of the late endosome. PLoS Pathog 2023; 19:e1011848. [PMID: 38055723 PMCID: PMC10727438 DOI: 10.1371/journal.ppat.1011848] [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: 10/17/2023] [Revised: 12/18/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023] Open
Abstract
Interaction between the Ebola virus envelope glycoprotein (GP) and the endosomal membrane is an essential step during virus entry into the cell. Acidic pH and Ca2+ have been implicated in mediating the GP-membrane interaction. However, the molecular mechanism by which these environmental factors regulate the conformational changes that enable engagement of GP with the target membrane is unknown. Here, we apply fluorescence correlation spectroscopy (FCS) and single-molecule Förster resonance energy transfer (smFRET) imaging to elucidate how the acidic pH, Ca2+ and anionic phospholipids in the late endosome promote GP-membrane interaction, thereby facilitating virus entry. We find that bis(monoacylglycero)phosphate (BMP), which is specific to the late endosome, is especially critical in determining the Ca2+-dependence of the GP-membrane interaction. Molecular dynamics (MD) simulations suggested residues in GP that sense pH and induce conformational changes that make the fusion loop available for insertion into the membrane. We similarly confirm residues in the fusion loop that mediate GP's interaction with Ca2+, which likely promotes local conformational changes in the fusion loop and mediates electrostatic interactions with the anionic phospholipids. Collectively, our results provide a mechanistic understanding of how the environment of the late endosome regulates the timing and efficiency of virus entry.
Collapse
Affiliation(s)
- Aastha Jain
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, Massachusetts, United States of America
| | - Ramesh Govindan
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, Massachusetts, United States of America
- Medical Scientist Training Program, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Alex R. Berkman
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, Massachusetts, United States of America
| | - Jeremy Luban
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, Massachusetts, United States of America
- Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, Massachusetts, United States of America
| | - Marco A. Díaz-Salinas
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, Massachusetts, United States of America
| | - Natasha D. Durham
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, Massachusetts, United States of America
| | - James B. Munro
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, Massachusetts, United States of America
- Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
24
|
Igarashi M, Hirokawa T, Takada A. Structural and Energetic Basis for Differential Binding of Ebola and Marburg Virus Glycoproteins to a Bat-Derived Niemann-Pick C1 Protein. J Infect Dis 2023; 228:S479-S487. [PMID: 37119290 DOI: 10.1093/infdis/jiad120] [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: 03/08/2023] [Revised: 04/14/2023] [Accepted: 04/26/2023] [Indexed: 05/01/2023] Open
Abstract
BACKGROUND Our previous study demonstrated that the fruit bat (Yaeyama flying fox)-derived cell line FBKT1 showed preferential susceptibility to Ebola virus (EBOV), whereas the human cell line HEK293T was similarly susceptible to EBOV and Marburg virus (MARV). This was due to 3 amino acid differences of the endosomal receptor Niemann-Pick C1 (NPC1) between FBKT1 and HEK293T (ie, TET and SGA, respectively, at positions 425-427), as well as 2 amino acid differences at positions 87 and 142 of the viral glycoprotein (GP) between EBOV and MARV. METHODS/RESULTS To understand the contribution of these amino acid differences to interactions between NPC1 and GP, we performed molecular dynamics simulations and binding free energy calculations. The average binding free energies of human NPC1 (hNPC1) and its mutant having TET at positions 425-427 (hNPC1/TET) were similar for the interaction with EBOV GP. In contrast, hNPC1/TET had a weaker interaction with MARV GP than wild-type hNPC1. As expected, substitutions of amino acid residues at 87 or 142 in EBOV and MARV GPs converted the binding affinity to hNPC1/TET. CONCLUSIONS Our data provide structural and energetic insights for understanding potential differences in the GP-NPC1 interaction, which could influence the host tropism of EBOV and MARV.
Collapse
Affiliation(s)
- Manabu Igarashi
- Division of Global Epidemiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Takatsugu Hirokawa
- Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Division of Biomedical Science, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Ayato Takada
- Division of Global Epidemiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
- Department of Disease Control, School of Veterinary Medicine, University of Zambia, Lusaka, Zambia
- One Health Research Center, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
25
|
Djurkovic MA, Leavitt CG, Arnett E, Kriachun V, Martínez-Sobrido L, Titone R, Sherwood LJ, Hayhurst A, Schlesinger LS, Shtanko O. Ebola Virus Uses Tunneling Nanotubes as an Alternate Route of Dissemination. J Infect Dis 2023; 228:S522-S535. [PMID: 37723997 PMCID: PMC10651192 DOI: 10.1093/infdis/jiad400] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023] Open
Abstract
Ebola virus (EBOV) disease is marked by rapid virus replication and spread. EBOV enters the cell by macropinocytosis and replicates in the cytoplasm, and nascent virions egress from the cell surface to infect neighboring cells. Here, we show that EBOV uses an alternate route to disseminate: tunneling nanotubes (TNTs). TNTs, an actin-based long-range intercellular communication system, allows for direct exchange of cytosolic constituents between cells. Using live, scanning electron, and high-resolution quantitative 3-dimensional microscopy, we show that EBOV infection of primary human cells results in the enhanced formation of TNTs containing viral nucleocapsids. TNTs promote the intercellular transfer of nucleocapsids in the absence of live virus, and virus could replicate in cells devoid of entry factors after initial stall. Our studies suggest an alternate model of EBOV dissemination within the host, laying the groundwork for further investigations into the pathogenesis of filoviruses and, importantly, stimulating new areas of antiviral design.
Collapse
Affiliation(s)
- Marija A Djurkovic
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio
| | - Carson G Leavitt
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio
| | - Eusondia Arnett
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio
| | - Valeriia Kriachun
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio
| | - Luis Martínez-Sobrido
- Disease Prevention and Intervention, Texas Biomedical Research Institute, San Antonio
| | - Rossella Titone
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio
| | - Laura J Sherwood
- Disease Prevention and Intervention, Texas Biomedical Research Institute, San Antonio
| | - Andrew Hayhurst
- Disease Prevention and Intervention, Texas Biomedical Research Institute, San Antonio
| | - Larry S Schlesinger
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio
| | - Olena Shtanko
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio
- Disease Prevention and Intervention, Texas Biomedical Research Institute, San Antonio
| |
Collapse
|
26
|
Jain A, Govindan R, Berkman A, Luban J, Durham ND, Munro J. Regulation of Ebola GP conformation and membrane binding by the chemical environment of the late endosome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.18.524651. [PMID: 36711925 PMCID: PMC9882366 DOI: 10.1101/2023.01.18.524651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Interaction between the Ebola virus envelope glycoprotein (GP) and the endosomal membrane is an essential step during virus entry into the cell. Acidic pH and Ca2+ have been implicated in mediating the GP-membrane interaction. However, the molecular mechanism by which these environmental factors regulate the conformational changes that enable engagement of GP with the target membrane is unknown. Here, we apply fluorescence correlation spectroscopy (FCS) and single-molecule Forster resonance energy transfer (smFRET) imaging to elucidate how the acidic pH, Ca2+ and anionic phospholipids in the late endosome promote GP-membrane interaction, thereby facilitating virus entry. We find that bis(monoacylglycero)phosphate (BMP), which is specific to the late endosome, is especially critical in determining the Ca2+-dependence of the GP-membrane interaction. Molecular dynamics (MD) simulations suggested residues in GP that sense pH and induce conformational changes that make the fusion loop available for insertion into the membrane. We similarly confirm residues in the fusion loop that mediate GPs interaction with Ca2+, which likely promotes local conformational changes in the fusion loop and mediates electrostatic interactions with the anionic phospholipids. Collectively, our results provide a mechanistic understanding of how the environment of the late endosome regulates the timing and efficiency of virus entry.
Collapse
|
27
|
Zhao S, Jiang M, Qing H, Ni J. Cathepsins and SARS-CoV-2 infection: From pathogenic factors to potential therapeutic targets. Br J Pharmacol 2023; 180:2455-2481. [PMID: 37403614 DOI: 10.1111/bph.16187] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/04/2023] [Accepted: 06/20/2023] [Indexed: 07/06/2023] Open
Abstract
Coronavirus disease-19 (COVID-19) is caused by severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection. The COVID-19 pandemic began in March 2020 and has wrought havoc on health and economic systems worldwide. Efficacious treatment for COVID-19 is lacking: Only preventive measures as well as symptomatic and supportive care are available. Preclinical and clinical studies have indicated that lysosomal cathepsins might contribute to the pathogenesis and disease outcome of COVID-19. Here, we discuss cutting-edge evidence on the pathological roles of cathepsins in SARS-CoV-2 infection, host immune dysregulations, and the possible underlying mechanisms. Cathepsins are attractive drug targets because of their defined substrate-binding pockets, which can be exploited as binding sites for pharmaceutical enzyme inhibitors. Accordingly, the potential modulatory strategies of cathepsin activity are discussed. These insights could shed light on the development of cathepsin-based interventions for COVID-19.
Collapse
Affiliation(s)
- Shuxuan Zhao
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Muzhou Jiang
- Department of Periodontics, Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
28
|
Gilbertson B, Subbarao K. What Have We Learned by Resurrecting the 1918 Influenza Virus? Annu Rev Virol 2023; 10:25-47. [PMID: 37774132 DOI: 10.1146/annurev-virology-111821-104408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2023]
Abstract
The 1918 Spanish influenza pandemic was one of the deadliest infectious disease events in recorded history, resulting in approximately 50-100 million deaths worldwide. The origins of the 1918 virus and the molecular basis for its exceptional virulence remained a mystery for much of the 20th century because the pandemic predated virologic techniques to isolate, passage, and store influenza viruses. In the late 1990s, overlapping fragments of influenza viral RNA preserved in the tissues of several 1918 victims were amplified and sequenced. The use of influenza reverse genetics then permitted scientists to reconstruct the 1918 virus entirely from cloned complementary DNA, leading to new insights into the origin of the virus and its pathogenicity. Here, we discuss some of the advances made by resurrection of the 1918 virus, including the rise of innovative molecular research, which is a topic in the dual use debate.
Collapse
Affiliation(s)
- Brad Gilbertson
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Victoria, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia;
| |
Collapse
|
29
|
Odongo L, Habtegebrael BH, Kiessling V, White JM, Tamm LK. A novel in vitro system of supported planar endosomal membranes (SPEMs) reveals an enhancing role for cathepsin B in the final stage of Ebola virus fusion and entry. Microbiol Spectr 2023; 11:e0190823. [PMID: 37728342 PMCID: PMC10581071 DOI: 10.1128/spectrum.01908-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/17/2023] [Indexed: 09/21/2023] Open
Abstract
Ebola virus (EBOV) causes a hemorrhagic fever with fatality rates up to 90%. The EBOV entry process is complex and incompletely understood. Following attachment to host cells, EBOV is trafficked to late endosomes/lysosomes where its glycoprotein (GP) is processed to a 19-kDa form, which binds to the EBOV intracellular receptor Niemann-Pick type C1. We previously showed that the cathepsin protease inhibitor, E-64d, blocks infection by pseudovirus particles bearing 19-kDa GP, suggesting that further cathepsin action is needed to trigger fusion. This, however, has not been demonstrated directly. Since 19-kDa Ebola GP fusion occurs in late endosomes, we devised a system in which enriched late endosomes are used to prepare supported planar endosomal membranes (SPEMs), and fusion of fluorescent (pseudo)virus particles is monitored by total internal reflection fluorescence microscopy. We validated the system by demonstrating the pH dependencies of influenza virus hemagglutinin (HA)-mediated and Lassa virus (LASV) GP-mediated fusion. Using SPEMs, we showed that fusion mediated by 19-kDa Ebola GP is dependent on low pH, enhanced by Ca2+, and augmented by the addition of cathepsins. Subsequently, we found that E-64d inhibits full fusion, but not lipid mixing, mediated by 19-kDa GP, which we corroborated with the reversible cathepsin inhibitor VBY-825. Hence, we provide both gain- and loss-of-function evidence that further cathepsin action enhances the fusion activity of 19-kDa Ebola GP. In addition to providing new insights into how Ebola GP mediates fusion, the approach we developed employing SPEMs can now be broadly used for studies of virus and toxin entry through endosomes. IMPORTANCE Ebola virus is the causative agent of Ebola virus disease, which is severe and frequently lethal. EBOV gains entry into cells via late endosomes/lysosomes. The events immediately preceding fusion of the viral and endosomal membranes are incompletely understood. In this study, we report a novel in vitro system for studying virus fusion with endosomal membranes. We validated the system by demonstrating the low pH dependencies of influenza and Lassa virus fusion. Moreover, we show that further cathepsin B action enhances the fusion activity of the primed Ebola virus glycoprotein. Finally, this model endosomal membrane system should be useful in studying the mechanisms of bilayer breaching by other enveloped viruses, by non-enveloped viruses, and by acid-activated bacterial toxins.
Collapse
Affiliation(s)
- Laura Odongo
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Betelihem H. Habtegebrael
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Volker Kiessling
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Judith M. White
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia, USA
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Lukas K. Tamm
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
30
|
Shepley-McTaggart A, Liang J, Ding Y, Djurkovic MA, Kriachun V, Shtanko O, Sunyer O, Harty RN. Contrasting effects of filamin A and B proteins in modulating filovirus entry. PLoS Pathog 2023; 19:e1011595. [PMID: 37585478 PMCID: PMC10461817 DOI: 10.1371/journal.ppat.1011595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 08/28/2023] [Accepted: 08/02/2023] [Indexed: 08/18/2023] Open
Abstract
Ebola (EBOV) and Marburg viruses (MARV) cause severe hemorrhagic fever associated with high mortality rates in humans. A better understanding of filovirus-host interactions that regulate the EBOV and MARV lifecycles can provide biological and mechanistic insight critical for therapeutic development. EBOV glycoprotein (eGP) and MARV glycoprotein (mGP) mediate entry into host cells primarily by actin-dependent macropinocytosis. Here, we identified actin-binding cytoskeletal crosslinking proteins filamin A (FLNa) and B (FLNb) as important regulators of both EBOV and MARV entry. We found that entry of pseudotype psVSV-RFP-eGP, infectious recombinant rVSV-eGP-mCherry, and live authentic EBOV and MARV was inhibited in filamin A knockdown (FLNaKD) cells, but was surprisingly enhanced in filamin B knockdown (FLNbKD) cells. Mechanistically, our findings suggest that differential regulation of macropinocytosis by FLNa and FLNb likely contributes to their specific effects on EBOV and MARV entry. This study is the first to identify the filamin family of proteins as regulators of EBOV and MARV entry. These findings may provide insight into the development of new countermeasures to prevent EBOV and MARV infections.
Collapse
Affiliation(s)
- Ariel Shepley-McTaggart
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jingjing Liang
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Yang Ding
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Marija A. Djurkovic
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Valeriia Kriachun
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Olena Shtanko
- Host-Pathogen Interactions, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Oriol Sunyer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ronald N. Harty
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
31
|
Liu D, Pan L, Zhai H, Qiu HJ, Sun Y. Virus tracking technologies and their applications in viral life cycle: research advances and future perspectives. Front Immunol 2023; 14:1204730. [PMID: 37334362 PMCID: PMC10272434 DOI: 10.3389/fimmu.2023.1204730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
Viruses are simple yet highly pathogenic microorganisms that parasitize within cells and pose serious threats to the health, economic development, and social stability of both humans and animals. Therefore, it is crucial to understand the dynamic mechanism of virus infection in hosts. One effective way to achieve this is through virus tracking technology, which utilizes fluorescence imaging to track the life processes of virus particles in living cells in real-time, providing a comprehensively and detailed spatiotemporal dynamic process and mechanism of virus infection. This paper provides a broad overview of virus tracking technology, including the selection of fluorescent labels and virus labeling components, the development of imaging microscopes, and its applications in various virus studies. Additionally, we discuss the possibilities and challenges of its future development, offering theoretical guidance and technical support for effective prevention and control of the viral disease outbreaks and epidemics.
Collapse
Affiliation(s)
| | | | | | - Hua-Ji Qiu
- *Correspondence: Hua-Ji Qiu, ; Yuan Sun,
| | - Yuan Sun
- *Correspondence: Hua-Ji Qiu, ; Yuan Sun,
| |
Collapse
|
32
|
Chen XN, Liang YF, Weng ZJ, Quan WP, Hu C, Peng YZ, Sun YS, Gao Q, Huang Z, Zhang GH, Gong L. Porcine Enteric Alphacoronavirus Entry through Multiple Pathways (Caveolae, Clathrin, and Macropinocytosis) Requires Rab GTPases for Endosomal Transport. J Virol 2023; 97:e0021023. [PMID: 36975780 PMCID: PMC10134835 DOI: 10.1128/jvi.00210-23] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/07/2023] [Indexed: 03/29/2023] Open
Abstract
Porcine enteric alphacoronavirus (PEAV) is a new bat HKU2-like porcine coronavirus, and its endemic outbreak has caused severe economic losses to the pig industry. Its broad cellular tropism suggests a potential risk of cross-species transmission. A limited understanding of PEAV entry mechanisms may hinder a rapid response to potential outbreaks. This study analyzed PEAV entry events using chemical inhibitors, RNA interference, and dominant-negative mutants. PEAV entry into Vero cells depended on three endocytic pathways: caveolae, clathrin, and macropinocytosis. Endocytosis requires dynamin, cholesterol, and a low pH. Rab5, Rab7, and Rab9 GTPases (but not Rab11) regulate PEAV endocytosis. PEAV particles colocalize with EEA1, Rab5, Rab7, Rab9, and Lamp-1, suggesting that PEAV translocates into early endosomes after internalization, and Rab5, Rab7, and Rab9 regulate trafficking to lysosomes before viral genome release. PEAV enters porcine intestinal cells (IPI-2I) through the same endocytic pathway, suggesting that PEAV may enter various cells through multiple endocytic pathways. This study provides new insights into the PEAV life cycle. IMPORTANCE Emerging and reemerging coronaviruses cause severe human and animal epidemics worldwide. PEAV is the first bat-like coronavirus to cause infection in domestic animals. However, the PEAV entry mechanism into host cells remains unknown. This study demonstrates that PEAV enters into Vero or IPI-2I cells through caveola/clathrin-mediated endocytosis and macropinocytosis, which does not require a specific receptor. Subsequently, Rab5, Rab7, and Rab9 regulate PEAV trafficking from early endosomes to lysosomes, which is pH dependent. The results advance our understanding of the disease and help to develop potential new drug targets against PEAV.
Collapse
Affiliation(s)
- Xiong-nan Chen
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan, People’s Republic of China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, People’s Republic of China
| | - Yi-fan Liang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, People’s Republic of China
| | - Zhi-jun Weng
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, People’s Republic of China
| | - Wei-peng Quan
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, People’s Republic of China
| | - Chen Hu
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
| | - Yun-zhao Peng
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, People’s Republic of China
| | - Ying-shuo Sun
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, People’s Republic of China
| | - Qi Gao
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, People’s Republic of China
| | - Zhao Huang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan, People’s Republic of China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, People’s Republic of China
| | - Gui-hong Zhang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, People’s Republic of China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, People’s Republic of China
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, People’s Republic of China
| | - Lang Gong
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, People’s Republic of China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong, People’s Republic of China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, People’s Republic of China
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, People’s Republic of China
| |
Collapse
|
33
|
Winter SL, Golani G, Lolicato F, Vallbracht M, Thiyagarajah K, Ahmed SS, Lüchtenborg C, Fackler OT, Brügger B, Hoenen T, Nickel W, Schwarz US, Chlanda P. The Ebola virus VP40 matrix layer undergoes endosomal disassembly essential for membrane fusion. EMBO J 2023:e113578. [PMID: 37082863 DOI: 10.15252/embj.2023113578] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 03/09/2023] [Accepted: 03/22/2023] [Indexed: 04/22/2023] Open
Abstract
Ebola viruses (EBOVs) assemble into filamentous virions, whose shape and stability are determined by the matrix viral protein 40 (VP40). Virus entry into host cells occurs via membrane fusion in late endosomes; however, the mechanism of how the remarkably long virions undergo uncoating, including virion disassembly and nucleocapsid release into the cytosol, remains unknown. Here, we investigate the structural architecture of EBOVs entering host cells and discover that the VP40 matrix disassembles prior to membrane fusion. We reveal that VP40 disassembly is caused by the weakening of VP40-lipid interactions driven by low endosomal pH that equilibrates passively across the viral envelope without a dedicated ion channel. We further show that viral membrane fusion depends on VP40 matrix integrity, and its disassembly reduces the energy barrier for fusion stalk formation. Thus, pH-driven structural remodeling of the VP40 matrix acts as a molecular switch coupling viral matrix uncoating to membrane fusion during EBOV entry.
Collapse
Affiliation(s)
- Sophie L Winter
- Schaller Research Groups, Department of Infectious Diseases, Virology, University Hospital Heidelberg, Heidelberg, Germany
- BioQuant-Center for Quantitative Biology, Heidelberg University, Heidelberg, Germany
| | - Gonen Golani
- BioQuant-Center for Quantitative Biology, Heidelberg University, Heidelberg, Germany
- Institute for Theoretical Physics, Heidelberg University, Heidelberg, Germany
| | - Fabio Lolicato
- Heidelberg University Biochemistry Center, Heidelberg, Germany
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Melina Vallbracht
- Schaller Research Groups, Department of Infectious Diseases, Virology, University Hospital Heidelberg, Heidelberg, Germany
- BioQuant-Center for Quantitative Biology, Heidelberg University, Heidelberg, Germany
| | - Keerthihan Thiyagarajah
- Schaller Research Groups, Department of Infectious Diseases, Virology, University Hospital Heidelberg, Heidelberg, Germany
- BioQuant-Center for Quantitative Biology, Heidelberg University, Heidelberg, Germany
| | - Samy Sid Ahmed
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Oliver T Fackler
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Britta Brügger
- Heidelberg University Biochemistry Center, Heidelberg, Germany
| | - Thomas Hoenen
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Insitut, Greifswald-Insel Riems, Greifswald, Germany
| | - Walter Nickel
- Heidelberg University Biochemistry Center, Heidelberg, Germany
| | - Ulrich S Schwarz
- BioQuant-Center for Quantitative Biology, Heidelberg University, Heidelberg, Germany
- Institute for Theoretical Physics, Heidelberg University, Heidelberg, Germany
| | - Petr Chlanda
- Schaller Research Groups, Department of Infectious Diseases, Virology, University Hospital Heidelberg, Heidelberg, Germany
- BioQuant-Center for Quantitative Biology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
34
|
Stewart CM, Bo Y, Fu K, Chan M, Kozak R, Apperley KYP, Laroche G, Daniel R, Beauchemin AM, Kobinger G, Kobasa D, Côté M. Sphingosine Kinases Promote Ebola Virus Infection and Can Be Targeted to Inhibit Filoviruses, Coronaviruses, and Arenaviruses Using Late Endocytic Trafficking to Enter Cells. ACS Infect Dis 2023; 9:1064-1077. [PMID: 37053583 DOI: 10.1021/acsinfecdis.2c00416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
Entry of enveloped viruses in host cells requires the fusion of viral and host cell membranes, a process that is facilitated by viral fusion proteins protruding from the viral envelope. These viral fusion proteins need to be triggered by host factors, and for some viruses, this event occurs inside endosomes and/or lysosomes. Consequently, these 'late-penetrating viruses' must be internalized and delivered to entry-conducive intracellular vesicles. Because endocytosis and vesicular trafficking are tightly regulated cellular processes, late-penetrating viruses also depend on specific host proteins for efficient delivery to the site of fusion, suggesting that these could be targeted for antiviral therapy. In this study, we investigated a role for sphingosine kinases (SKs) in viral entry and found that chemical inhibition of sphingosine kinase 1 (SK1) and/or SK2 and knockdown of SK1/2 inhibited entry of Ebola virus (EBOV) into host cells. Mechanistically, inhibition of SK1/2 prevented EBOV from reaching late-endosomes and lysosomes that contain the EBOV receptor, Niemann Pick C1 (NPC1). Furthermore, we present evidence that suggests that the trafficking defect caused by SK1/2 inhibition occurs independently of sphingosine-1-phosphate (S1P) signaling through cell-surface S1P receptors. Lastly, we found that chemical inhibition of SK1/2 prevents entry of other late-penetrating viruses, including arenaviruses and coronaviruses, and inhibits infection by replication-competent EBOV and SARS-CoV-2 in Huh7.5 cells. In sum, our results highlight an important role played by SK1/2 in endocytic trafficking, which can be targeted to inhibit entry of late-penetrating viruses and could serve as a starting point for the development of broad-spectrum antiviral therapeutics.
Collapse
Affiliation(s)
- Corina M Stewart
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Yuxia Bo
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Kathy Fu
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Mable Chan
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
- Department of Infectious Diseases and Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Robert Kozak
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
| | - Kim Yang-Ping Apperley
- Center for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Geneviève Laroche
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Redaet Daniel
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - André M Beauchemin
- Center for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Gary Kobinger
- Galveston National Laboratory, Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas 77550, United States
| | - Darwyn Kobasa
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
- Department of Infectious Diseases and Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Center for Catalysis Research and Innovation, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| |
Collapse
|
35
|
Mardi N, Haiaty S, Rahbarghazi R, Mobarak H, Milani M, Zarebkohan A, Nouri M. Exosomal transmission of viruses, a two-edged biological sword. Cell Commun Signal 2023; 21:19. [PMID: 36691072 PMCID: PMC9868521 DOI: 10.1186/s12964-022-01037-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 12/28/2022] [Indexed: 01/24/2023] Open
Abstract
As a common belief, most viruses can egress from the host cells as single particles and transmit to uninfected cells. Emerging data have revealed en bloc viral transmission as lipid bilayer-cloaked particles via extracellular vesicles especially exosomes (Exo). The supporting membrane can be originated from multivesicular bodies during intra-luminal vesicle formation and autophagic response. Exo are nano-sized particles, ranging from 40-200 nm, with the ability to harbor several types of signaling molecules from donor to acceptor cells in a paracrine manner, resulting in the modulation of specific signaling reactions in target cells. The phenomenon of Exo biogenesis consists of multiple and complex biological steps with the participation of diverse constituents and molecular pathways. Due to similarities between Exo biogenesis and virus replication and the existence of shared pathways, it is thought that viruses can hijack the Exo biogenesis machinery to spread and evade immune cells. To this end, Exo can transmit complete virions (as single units or aggregates), separate viral components, and naked genetic materials. The current review article aims to scrutinize challenges and opportunities related to the exosomal delivery of viruses in terms of viral infections and public health. Video Abstract.
Collapse
Affiliation(s)
- Narges Mardi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanya Haiaty
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Halimeh Mobarak
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, Iran
| | - Morteza Milani
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zarebkohan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
36
|
Chen T, Tu S, Ding L, Jin M, Chen H, Zhou H. The role of autophagy in viral infections. J Biomed Sci 2023; 30:5. [PMID: 36653801 PMCID: PMC9846652 DOI: 10.1186/s12929-023-00899-2] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/10/2023] [Indexed: 01/20/2023] Open
Abstract
Autophagy is an evolutionarily conserved catabolic cellular process that exerts antiviral functions during a viral invasion. However, co-evolution and co-adaptation between viruses and autophagy have armed viruses with multiple strategies to subvert the autophagic machinery and counteract cellular antiviral responses. Specifically, the host cell quickly initiates the autophagy to degrade virus particles or virus components upon a viral infection, while cooperating with anti-viral interferon response to inhibit the virus replication. Degraded virus-derived antigens can be presented to T lymphocytes to orchestrate the adaptive immune response. Nevertheless, some viruses have evolved the ability to inhibit autophagy in order to evade degradation and immune responses. Others induce autophagy, but then hijack autophagosomes as a replication site, or hijack the secretion autophagy pathway to promote maturation and egress of virus particles, thereby increasing replication and transmission efficiency. Interestingly, different viruses have unique strategies to counteract different types of selective autophagy, such as exploiting autophagy to regulate organelle degradation, metabolic processes, and immune responses. In short, this review focuses on the interaction between autophagy and viruses, explaining how autophagy serves multiple roles in viral infection, with either proviral or antiviral functions.
Collapse
Affiliation(s)
- Tong Chen
- grid.35155.370000 0004 1790 4137State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430030 China ,grid.35155.370000 0004 1790 4137Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430030 China
| | - Shaoyu Tu
- grid.35155.370000 0004 1790 4137State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430030 China ,grid.35155.370000 0004 1790 4137Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430030 China
| | - Ling Ding
- grid.35155.370000 0004 1790 4137State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430030 China ,grid.35155.370000 0004 1790 4137Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430030 China
| | - Meilin Jin
- grid.35155.370000 0004 1790 4137State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430030 China ,grid.35155.370000 0004 1790 4137Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430030 China
| | - Huanchun Chen
- grid.35155.370000 0004 1790 4137State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430030 China ,grid.35155.370000 0004 1790 4137Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430030 China
| | - Hongbo Zhou
- grid.35155.370000 0004 1790 4137State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430030 China ,grid.35155.370000 0004 1790 4137Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430030 China
| |
Collapse
|
37
|
Khwaza V, Buyana B, Nqoro X, Peter S, Mbese Z, Feketshane Z, Alven S, Aderibigbe BA. Strategies for delivery of antiviral agents. VIRAL INFECTIONS AND ANTIVIRAL THERAPIES 2023:407-492. [DOI: 10.1016/b978-0-323-91814-5.00018-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
38
|
Seneca Valley Virus Enters PK-15 Cells via Caveolae-Mediated Endocytosis and Macropinocytosis Dependent on Low-pH, Dynamin, Rab5, and Rab7. J Virol 2022; 96:e0144622. [PMID: 36472440 PMCID: PMC9769397 DOI: 10.1128/jvi.01446-22] [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: 12/12/2022] Open
Abstract
Seneca Valley virus (SVV), a new pathogen resulting in porcine vesicular disease, is prevalent in pig herds worldwide. Although an understanding of SVV biology pathogenesis is crucial for preventing and controlling this disease, the molecular mechanisms for the entry and post-internalization of SVV, which represent crucial steps in viral infection, are not well characterized. In this study, specific inhibitors, Western blotting, and immunofluorescence detection revealed that SVV entry into PK-15 cells depends on low-pH conditions and dynamin. Furthermore, results showed that caveolae-mediated endocytosis (CavME) contributes crucially to the internalization of SVV, as evidenced by cholesterol depletion, downregulation of caveolin-1 expression by small interfering RNA knockdown, and overexpression of a caveolin-1 dominant negative (caveolin-1-DN) in SVV-infected PK-15 cells. However, SVV entry into PK-15 cells did not depend on clathrin-mediated endocytosis (CME). Furthermore, treatment with specific inhibitors demonstrated that SVV entry into PK-15 cells via macropinocytosis depended on the Na+/H+ exchanger (NHE), p21-activated kinase 1 (Pak1), and actin rearrangement, but not phosphatidylinositol 3-kinase (PI3K). Electron microscopy showed that SVV particles or proteins were localized in CavME and macropinocytosis. Finally, knockdown of GTPase Rab5 and Rab7 by siRNA significantly inhibited SVV replication, as determined by measuring viral genome copy numbers, viral protein expression, and viral titers. In this study, our results demonstrated that SVV utilizes caveolae-mediated endocytosis and macropinocytosis to enter PK-15 cells, dependent on low pH, dynamin, Rab5, and Rab7. IMPORTANCE Entry of virus into cells represents the initiation of a successful infection. As an emerging pathogen of porcine vesicular disease, clarification of the process of SVV entry into cells enables us to better understand the viral life cycle and pathogenesis. In this study, patterns of SVV internalization and key factors required were explored. We demonstrated for the first time that SVV entry into PK-15 cells via caveolae-mediated endocytosis and macropinocytosis requires Rab5 and Rab7 and is independent of clathrin-mediated endocytosis, and that low-pH conditions and dynamin are involved in the process of SVV internalization. This information increases our understanding of the patterns in which all members of the family Picornaviridae enter host cells, and provides new insights for preventing and controlling SVV infection.
Collapse
|
39
|
Yi D, Li Q, Wang H, Lv K, Ma L, Wang Y, Wang J, Zhang Y, Liu M, Li X, Qi J, Shi Y, Gao GF, Cen S. Repurposing of berbamine hydrochloride to inhibit Ebola virus by targeting viral glycoprotein. Acta Pharm Sin B 2022; 12:4378-4389. [PMID: 36561997 PMCID: PMC9764067 DOI: 10.1016/j.apsb.2022.05.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/07/2022] [Accepted: 05/12/2022] [Indexed: 12/25/2022] Open
Abstract
Ebola virus (EBOV) infection leads to staggeringly high mortality rate. Effective and low-cost treatments are urgently needed to control frequent EBOV outbreaks in Africa. In this study, we report that a natural compound called berbamine hydrochloride strongly inhibits EBOV replication in vitro and in vivo. Our work further showed that berbamine hydrochloride acts by directly binding to the cleaved EBOV glycoprotein (GPcl), disrupting GPcl interaction with viral receptor Niemann-Pick C1, thus blocking the fusion of viral and cellular membranes. Our data support the probability of developing anti-EBOV small molecule drugs by targeting viral GPcl. More importantly, since berbamine hydrochloride has been used in clinic to treat leukopenia, it holds great promise of being quickly repurposed as an anti-EBOV drug.
Collapse
Affiliation(s)
- Dongrong Yi
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing 100050, China
| | - Quanjie Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing 100050, China
| | - Han Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Kai Lv
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing 100050, China
| | - Ling Ma
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing 100050, China
| | - Yujia Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing 100050, China
| | - Jing Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing 100050, China
| | - Yongxin Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing 100050, China
| | - Mingliang Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing 100050, China
| | - Xiaoyu Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing 100050, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Shi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China,University of Chinese Academy of Sciences, Beijing 100049, China,Corresponding authors.
| | - George F. Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shan Cen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical School, Beijing 100050, China,CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100050, China,Corresponding authors.
| |
Collapse
|
40
|
Lu J, Gullett JM, Kanneganti TD. Filoviruses: Innate Immunity, Inflammatory Cell Death, and Cytokines. Pathogens 2022; 11:1400. [PMID: 36558734 PMCID: PMC9785368 DOI: 10.3390/pathogens11121400] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
Filoviruses are a group of single-stranded negative sense RNA viruses. The most well-known filoviruses that affect humans are ebolaviruses and marburgviruses. During infection, they can cause life-threatening symptoms such as inflammation, tissue damage, and hemorrhagic fever, with case fatality rates as high as 90%. The innate immune system is the first line of defense against pathogenic insults such as filoviruses. Pattern recognition receptors (PRRs), including toll-like receptors, retinoic acid-inducible gene-I-like receptors, C-type lectin receptors, AIM2-like receptors, and NOD-like receptors, detect pathogens and activate downstream signaling to induce the production of proinflammatory cytokines and interferons, alert the surrounding cells to the threat, and clear infected and damaged cells through innate immune cell death. However, filoviruses can modulate the host inflammatory response and innate immune cell death, causing an aberrant immune reaction. Here, we discuss how the innate immune system senses invading filoviruses and how these deadly pathogens interfere with the immune response. Furthermore, we highlight the experimental difficulties of studying filoviruses as well as the current state of filovirus-targeting therapeutics.
Collapse
|
41
|
Husby ML, Amiar S, Prugar LI, David EA, Plescia CB, Huie KE, Brannan JM, Dye JM, Pienaar E, Stahelin RV. Phosphatidylserine clustering by the Ebola virus matrix protein is a critical step in viral budding. EMBO Rep 2022; 23:e51709. [PMID: 36094794 PMCID: PMC9638875 DOI: 10.15252/embr.202051709] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 07/28/2023] Open
Abstract
Phosphatidylserine (PS) is a critical lipid factor in the assembly and spread of numerous lipid-enveloped viruses. Here, we describe the ability of the Ebola virus (EBOV) matrix protein eVP40 to induce clustering of PS and promote viral budding in vitro, as well as the ability of an FDA-approved drug, fendiline, to reduce PS clustering and subsequent virus budding and entry. To gain mechanistic insight into fendiline inhibition of EBOV replication, multiple in vitro assays were run including imaging, viral budding and viral entry assays. Fendiline lowers PS content in mammalian cells and PS in the plasma membrane, where the ability of VP40 to form new virus particles is greatly lower. Further, particles that form from fendiline-treated cells have altered particle morphology and cannot significantly infect/enter cells. These complementary studies reveal the mechanism by which EBOV matrix protein clusters PS to enhance viral assembly, budding, and spread from the host cell while also laying the groundwork for fundamental drug targeting strategies.
Collapse
Affiliation(s)
- Monica L Husby
- Department of Medicinal Chemistry & Molecular PharmacologyPurdue UniversityWest LafayetteINUSA
- Purdue Institute of Inflammation, Immunology and Infectious Disease (PI4D)Purdue University, West LafayetteWest LafayetteINUSA
| | - Souad Amiar
- Department of Medicinal Chemistry & Molecular PharmacologyPurdue UniversityWest LafayetteINUSA
- Purdue Institute of Inflammation, Immunology and Infectious Disease (PI4D)Purdue University, West LafayetteWest LafayetteINUSA
| | - Laura I Prugar
- United States Army Medical Research Institute of Infectious Diseases USAMRIIDFort DetrickFrederickMDUSA
| | - Emily A David
- Department of Medicinal Chemistry & Molecular PharmacologyPurdue UniversityWest LafayetteINUSA
| | - Caroline B Plescia
- Department of Medicinal Chemistry & Molecular PharmacologyPurdue UniversityWest LafayetteINUSA
| | - Kathleen E Huie
- United States Army Medical Research Institute of Infectious Diseases USAMRIIDFort DetrickFrederickMDUSA
| | - Jennifer M Brannan
- United States Army Medical Research Institute of Infectious Diseases USAMRIIDFort DetrickFrederickMDUSA
| | - John M Dye
- United States Army Medical Research Institute of Infectious Diseases USAMRIIDFort DetrickFrederickMDUSA
| | - Elsje Pienaar
- Purdue Institute of Inflammation, Immunology and Infectious Disease (PI4D)Purdue University, West LafayetteWest LafayetteINUSA
- Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteINUSA
| | - Robert V Stahelin
- Department of Medicinal Chemistry & Molecular PharmacologyPurdue UniversityWest LafayetteINUSA
- Purdue Institute of Inflammation, Immunology and Infectious Disease (PI4D)Purdue University, West LafayetteWest LafayetteINUSA
| |
Collapse
|
42
|
Furuyama W, Sakaguchi M, Yamada K, Nanbo A. Development of an imaging system for visualization of Ebola virus glycoprotein throughout the viral lifecycle. Front Microbiol 2022; 13:1026644. [PMID: 36406413 PMCID: PMC9669576 DOI: 10.3389/fmicb.2022.1026644] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/16/2022] [Indexed: 11/06/2022] Open
Abstract
Ebola virus (EBOV) causes severe EBOV disease (EVD) in humans and non-human primates. Currently, limited countermeasures are available, and the virus must be studied in biosafety level-4 (BSL-4) laboratories. EBOV glycoprotein (GP) is a single transmembrane protein responsible for entry into host cells and is the target of multiple approved drugs. However, the molecular mechanisms underlying the intracellular dynamics of GP during EBOV lifecycle are poorly understood. In this study, we developed a novel GP monitoring system using transcription- and replication-competent virus-like particles (trVLPs) that enables the modeling of the EBOV lifecycle under BSL-2 conditions. We constructed plasmids to generate trVLPs containing the coding sequence of EBOV GP, in which the mucin-like domain (MLD) was replaced with fluorescent proteins. The generated trVLP efficiently replicated over multiple generations was similar to the wild type trVLP. Furthermore, we confirmed that the novel trVLP system enabled real-time visualization of GP throughout the trVLP replication cycle and exhibited intracellular localization similar to that of wild type GP. In summary, this novel monitoring system for GP will enable the characterization of the molecular mechanism of the EBOV lifecycle and can be applied for the development of therapeutics against EVD.
Collapse
Affiliation(s)
- Wakako Furuyama
- Department of Virus Infection Dynamics, National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki, Japan
| | - Miako Sakaguchi
- Central Laboratory, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Kento Yamada
- Department of Virus Infection Dynamics, National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki, Japan
| | - Asuka Nanbo
- Department of Virus Infection Dynamics, National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki, Japan
- *Correspondence: Asuka Nanbo,
| |
Collapse
|
43
|
Maekawa M, Natsume R, Arita M. Functional significance of ion channels during macropinosome resolution in immune cells. Front Physiol 2022; 13:1037758. [DOI: 10.3389/fphys.2022.1037758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022] Open
Abstract
Macropinocytosis is a unique type of endocytosis accompanied by membrane ruffle formation. Closure of membrane ruffles leads to the uptake of large volumes of fluid phase and, subsequently, the formation of large vacuoles termed macropinosomes. Immune cells, such as dendritic cells, T cells, and macrophages, endocytose the surrounding amino acids and pathogens via macropinocytosis either constitutively or in a stimulus-dependent fashion. This process is critical for cell migration, mammalian target of rapamycin complex 1 (mTORC1) activation, and antigen presentation. Large vacuoles are fragmented into tubules and smaller vesicles during the progression and maturation of macropinosomes in immune cells. This process is called “macropinosome resolution” and requires osmotically driven shrinkage of macropinosomes, which is controlled by ion channels present in them. The crenation of membranes on shrunken macropinosomes is recognized by curvature-sensing proteins and results in intracellular membrane trafficking. In this mini review, we highlight the recent progress in research on macropinosome resolution in macrophages, with a focus on ion channels (TPC1/2 for Na+ and TMEM206 for Cl−) that is required for macropinosome resolution. We also discuss the potential contribution of membrane lipids to this process.
Collapse
|
44
|
SARS-CoV-2 hijacks macropinocytosis to facilitate its entry and promote viral spike-mediated cell-to-cell fusion. J Biol Chem 2022; 298:102511. [PMID: 36259516 PMCID: PMC9484108 DOI: 10.1016/j.jbc.2022.102511] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 12/03/2022] Open
Abstract
Revealing the mechanisms of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) entry and cell-to-cell spread might provide insights for understanding the underlying mechanisms of viral pathogenesis, tropism, and virulence. The signaling pathways involved in SARS-CoV-2 entry and viral spike–mediated cell-to-cell fusion remain elusive. In the current study, we found that macropinocytosis inhibitors significantly suppressed SARS-CoV-2 infection at both the entry and viral spike–mediated cell-to-cell fusion steps. We demonstrated that SARS-CoV-2 entry required the small GTPase Rac1 and its effector kinase p21-activated kinase 1 by dominant-negative and RNAi assays in human embryonic kidney 293T–angiotensin-converting enzyme 2 cells and that the serine protease transmembrane serine protease 2 reversed the decrease in SARS-CoV-2 entry caused by the macropinocytosis inhibitors. Moreover, in the cell-to-cell fusion assay, we confirmed that macropinocytosis inhibitors significantly decreased viral spike–mediated cell-to-cell fusion. Overall, we provided evidence that SARS-CoV-2 utilizes a macropinocytosis pathway to enter target cells and to efficiently promote viral spike–mediated cell-to-cell fusion.
Collapse
|
45
|
CAPG Is Required for Ebola Virus Infection by Controlling Virus Egress from Infected Cells. Viruses 2022; 14:v14091903. [PMID: 36146710 PMCID: PMC9505868 DOI: 10.3390/v14091903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
The replication of Ebola virus (EBOV) is dependent upon actin functionality, especially at cell entry through macropinocytosis and at release of virus from cells. Previously, major actin-regulatory factors involved in actin nucleation, such as Rac1 and Arp2/3, were shown important in both steps. However, downstream of nucleation, many other cell factors are needed to control actin dynamics. How these regulate EBOV infection remains largely unclear. Here, we identified the actin-regulating protein, CAPG, as important for EBOV replication. Notably, knockdown of CAPG specifically inhibited viral infectivity and yield of infectious particles. Cell-based mechanistic analysis revealed a requirement of CAPG for virus production from infected cells. Proximity ligation and split-green fluorescent protein reconstitution assays revealed strong association of CAPG with VP40 that was mediated through the S1 domain of CAPG. Overall, CAPG is a novel host factor regulating EBOV infection through connecting actin filament stabilization to viral egress from cells.
Collapse
|
46
|
Peng W, Rayaprolu V, Parvate AD, Pronker MF, Hui S, Parekh D, Shaffer K, Yu X, Saphire EO, Snijder J. Glycan shield of the ebolavirus envelope glycoprotein GP. Commun Biol 2022; 5:785. [PMID: 35927436 PMCID: PMC9352669 DOI: 10.1038/s42003-022-03767-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 07/25/2022] [Indexed: 11/09/2022] Open
Abstract
The envelope glycoprotein GP of the ebolaviruses is essential for host cell entry and the primary target of the host antibody response. GP is heavily glycosylated with up to 17 N-linked sites, numerous O-linked glycans in its disordered mucin-like domain (MLD), and three predicted C-linked mannosylation sites. Glycosylation is important for host cell attachment, GP stability and fusion activity, and shielding from neutralization by serum antibodies. Here, we use glycoproteomics to profile the site-specific glycosylation patterns of ebolavirus GP. We detect up to 16 unique O-linked glycosylation sites in the MLD, and two O-linked sites in the receptor-binding GP1 subunit. Multiple O-linked glycans are observed within N-linked glycosylation sequons, suggesting crosstalk between the two types of modifications. We confirmed C-mannosylation of W288 in full-length trimeric GP. We find complex glycosylation at the majority of N-linked sites, while the conserved sites N257 and especially N563 are enriched in unprocessed glycans, suggesting a role in host-cell attachment via DC-SIGN/L-SIGN. Our findings illustrate how N-, O-, and C-linked glycans together build the heterogeneous glycan shield of GP, guiding future immunological studies and functional interpretation of ebolavirus GP-antibody interactions. Site-specific N-, O-, and C-linked glycans are characterized in the ebolavirus envelope glycoprotein GP using mass spectrometry-based glycoproteomics.
Collapse
Affiliation(s)
- Weiwei Peng
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584, CH, Utrecht, The Netherlands
| | - Vamseedhar Rayaprolu
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.,Pacific Northwest Center for CryoEM, Portland, OR, 97225, USA
| | - Amar D Parvate
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.,Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Matti F Pronker
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584, CH, Utrecht, The Netherlands
| | - Sean Hui
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.,Molecular Microbiology and Microbial Pathogenesis Program, Washington University School of Medicine, Saint Louis, MO, 63108, USA
| | - Diptiben Parekh
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Kelly Shaffer
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Xiaoying Yu
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Erica O Saphire
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.,Department of Medicine, University of California, San Diego, La Jolla, CA, 92039, USA
| | - Joost Snijder
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584, CH, Utrecht, The Netherlands.
| |
Collapse
|
47
|
Abstract
Filovirus-infected cells are characterized by typical cytoplasmic inclusion bodies (IBs) located in the perinuclear region. The formation of these IBs is induced mainly by the accumulation of the filoviral nucleoprotein NP, which recruits the other nucleocapsid proteins, the polymerase co-factor VP35, the polymerase L, the transcription factor VP30 and VP24 via direct or indirect protein-protein interactions. Replication of the negative-strand RNA genomes by the viral polymerase L and VP35 occurs in the IBs, resulting in the synthesis of positive-strand genomes, which are encapsidated by NP, thus forming ribonucleoprotein complexes (antigenomic RNPs). These newly formed antigenomic RNPs in turn serve as templates for the synthesis of negative-strand RNA genomes that are also encapsidated by NP (genomic RNPs). Still in the IBs, genomic RNPs mature into tightly packed transport-competent nucleocapsids (NCs) by the recruitment of the viral protein VP24. NCs are tightly coiled left-handed helices whose structure is mainly determined by the multimerization of NP at its N-terminus, and these helices form the inner layer of the NCs. The RNA genome is fixed by 2 lobes of the NP N-terminus and is thus guided by individual NP molecules along the turns of the helix. Direct interaction of the NP C-terminus with the VP35 and VP24 molecules forms the outer layer of the NCs. Once formed, NCs that are located at the border of the IBs recruit actin polymerization machinery to one of their ends to drive their transport to budding sites for their envelopment and final release. Here, we review the current knowledge on the structure, assembly, and transport of filovirus NCs.
Collapse
Affiliation(s)
- Olga Dolnik
- Institute of Virology, Philipps-University Marburg, Marburg, Germany
| | - Stephan Becker
- Institute of Virology, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
48
|
Fujioka Y, Kashiwagi S, Yoshida A, Satoh AO, Fujioka M, Amano M, Yamauchi Y, Ohba Y. A method for the generation of pseudovirus particles bearing SARS coronavirus spike protein in high yields. Cell Struct Funct 2022; 47:43-53. [PMID: 35491102 PMCID: PMC10511058 DOI: 10.1247/csf.21047] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 04/19/2022] [Indexed: 12/17/2023] Open
Abstract
The ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has threatened human health and the global economy. Development of additional vaccines and therapeutics is urgently required, but such development with live virus must be conducted with biosafety level 3 confinement. Pseudotyped viruses have been widely adopted for studies of virus entry and pharmaceutical development to overcome this restriction. Here we describe a modified protocol to generate vesicular stomatitis virus (VSV) pseudotyped with SARS-CoV or SARS-CoV-2 spike protein in high yield. We found that a large proportion of pseudovirions produced with the conventional transient expression system lacked coronavirus spike protein at their surface as a result of inhibition of parental VSV infection by overexpression of this protein. Establishment of stable cell lines with an optimal expression level of coronavirus spike protein allowed the efficient production of progeny pseudoviruses decorated with spike protein. This improved VSV pseudovirus production method should facilitate studies of coronavirus entry and development of antiviral agents.Key words: severe acute respiratory syndrome coronavirus (SARS-CoV), SARS-CoV-2, pseudovirus, vesicular stomatitis virus (VSV), spike protein.
Collapse
Affiliation(s)
- Yoichiro Fujioka
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, N12W6, Kita-ku, Sapporo 060-8612, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, N15W7, Kita-ku, Sapporo 060-8638, Japan
| | - Sayaka Kashiwagi
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, N12W6, Kita-ku, Sapporo 060-8612, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, N15W7, Kita-ku, Sapporo 060-8638, Japan
| | - Aiko Yoshida
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, N12W6, Kita-ku, Sapporo 060-8612, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, N15W7, Kita-ku, Sapporo 060-8638, Japan
| | - Aya O. Satoh
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, N15W7, Kita-ku, Sapporo 060-8638, Japan
| | - Mari Fujioka
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, N15W7, Kita-ku, Sapporo 060-8638, Japan
| | - Maho Amano
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, N15W7, Kita-ku, Sapporo 060-8638, Japan
| | - Yohei Yamauchi
- AMED-CREST, Japan Agency for Medical Research and Development, N15W7, Kita-ku, Sapporo 060-8638, Japan
- School of Cellular and Molecular Medicine, University of Bristol, University Walk, Bristol BS8 1TD, UK
- Division of Biological Science, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan
| | - Yusuke Ohba
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15W7, Kita-ku, Sapporo 060-8638, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, N12W6, Kita-ku, Sapporo 060-8612, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, N15W7, Kita-ku, Sapporo 060-8638, Japan
| |
Collapse
|
49
|
Plescia CB, Lindstrom AR, Quintero MV, Keiser P, Anantpadma M, Davey R, Stahelin RV, Davisson VJ. Evaluation of Phenol-Substituted Diphyllin Derivatives as Selective Antagonists for Ebola Virus Entry. ACS Infect Dis 2022; 8:942-957. [PMID: 35357134 PMCID: PMC9112336 DOI: 10.1021/acsinfecdis.1c00474] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Ebola
virus (EBOV) is an aggressive filoviral pathogen that can
induce severe hemorrhagic fever in humans with up to 90% fatality
rate. To date, there are no clinically effective small-molecule drugs
for postexposure therapies to treat filoviral infections. EBOV cellular
entry and infection involve uptake via macropinocytosis, navigation
through the endocytic pathway, and pH-dependent escape into the cytoplasm.
We report the inhibition of EBOV cell entry via selective inhibition
of vacuolar (V)-ATPase by a new series of phenol-substituted derivatives
of the natural product scaffold diphyllin. In cells challenged with
Ebola virus, the diphyllin derivatives inhibit viral entry dependent
upon structural variations to low nanomolar potencies. Mechanistically,
the diphyllin derivatives had no effect on uptake and colocalization
of viral particles with endocytic marker LAMP1 but directly modulated
endosomal pH. The most potent effects were reversible exhibiting higher
selectivity than bafilomycin or the parent diphyllin. Unlike general
lysosomotrophic agents, the diphyllin derivatives showed no major
disruptions of endocytic populations or morphology when examined with
Rab5 and LAMP1 markers. The dilated vacuole phenotype induced by apilimod
treatment or in constitutively active Rab5 mutant Q79L-expressing
cells was both blocked and reversed by the diphyllin derivatives.
The results are consistent with the action of the diphyllin scaffold
as a selective pH-dependent viral entry block in late endosomes. Overall,
the compounds show improved selectivity and minimal cytotoxicity relative
to classical endosomal acidification blocking agents.
Collapse
Affiliation(s)
| | | | - Maritza V. Quintero
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio 78229-3900, United States
| | - Patrick Keiser
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts 02118, United States
| | - Manu Anantpadma
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts 02118, United States
| | - Robert Davey
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts 02118, United States
| | | | | |
Collapse
|
50
|
Unique Mode of Antiviral Action of a Marine Alkaloid against Ebola Virus and SARS-CoV-2. Viruses 2022; 14:v14040816. [PMID: 35458549 PMCID: PMC9028129 DOI: 10.3390/v14040816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/24/2022] [Accepted: 04/02/2022] [Indexed: 11/16/2022] Open
Abstract
Lamellarin α 20-sulfate is a cell-impenetrable marine alkaloid that can suppress infection that is mediated by the envelope glycoprotein of human immunodeficiency virus type 1. We explored the antiviral action and mechanisms of this alkaloid against emerging enveloped RNA viruses that use endocytosis for infection. The alkaloid inhibited the infection of retroviral vectors that had been pseudotyped with the envelope glycoprotein of Ebola virus and SARS-CoV-2. The antiviral effects of lamellarin were independent of the retrovirus Gag-Pol proteins. Interestingly, although heparin and dextran sulfate suppressed the cell attachment of vector particles, lamellarin did not. In silico structural analyses of the trimeric glycoprotein of the Ebola virus disclosed that the principal lamellarin-binding site is confined to a previously unappreciated cavity near the NPC1-binding site and fusion loop, whereas those for heparin and dextran sulfate were dispersed across the attachment and fusion subunits of the glycoproteins. Notably, lamellarin binding to this cavity was augmented under conditions where the pH was 5.0. These results suggest that the final action of the alkaloid against Ebola virus is specific to events following endocytosis, possibly during conformational glycoprotein changes in the acidic environment of endosomes. Our findings highlight the unique biological and physicochemical features of lamellarin α 20-sulfate and should lead to the further use of broadly reactive antivirals to explore the structural mechanisms of virus replication.
Collapse
|