1
|
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
|
2
|
Wang LL, Alfson K, Eaton B, Mattix ME, Goez-Gazi Y, Holbrook MR, Carrion R, Xiang SH. Algal Lectin Griffithsin Inhibits Ebola Virus Infection. Molecules 2025; 30:892. [PMID: 40005201 PMCID: PMC11858388 DOI: 10.3390/molecules30040892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Algal lectin Griffithsin (GRFT) is a well-known mannose-binding protein which has broad-spectrum antiviral activity against several important infectious viruses including HIV, HCV, and SARS-CoV-2. Therefore, GRFT has been brought great attention to antiviral therapeutic development. In this report, we have tested GRFT's activity against the lethal Ebola virus in vitro and in vivo. Our data have shown that the IC50 value is about 42 nM for inhibiting Zaire Ebola virus (EBOV) infection in vitro. The preliminary in vivo mice model using mouse-adapted EBOV has also shown a certain efficacy for delayed mortality compared to the control animals. A GRFT pull-down experiment using viral particles demonstrates that GRFT can bind to N-glycans of EBOV. Thus, it can be concluded that GRFT, through binding to viral glycans, may block Ebola virus infection and has potential for the treatment of Ebola virus disease (EVD).
Collapse
Affiliation(s)
- Leah Liu Wang
- Nebraska Center for Virology, School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, USA;
| | - Kendra Alfson
- Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA (Y.G.-G.)
| | - Brett Eaton
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, Frederick, MD 21702, USA
| | - Marc E. Mattix
- Nonclinical Pathology Services, LLC, 5920 Clubhouse Pointe Dr., Medina 44256, OH, USA
| | - Yenny Goez-Gazi
- Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA (Y.G.-G.)
| | - Michael R. Holbrook
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, Frederick, MD 21702, USA
| | - Ricardo Carrion
- Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA (Y.G.-G.)
| | - Shi-Hua Xiang
- Nebraska Center for Virology, School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68588, USA;
| |
Collapse
|
3
|
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
|
4
|
Chicano Wust I. Viral interactions with host factors (TIM-1, TAM -receptors, Glut-1) are related to the disruption of glucose and ascorbate transport and homeostasis, causing the haemorrhagic manifestations of viral haemorrhagic fevers. F1000Res 2024; 12:518. [PMID: 39931159 PMCID: PMC11809632 DOI: 10.12688/f1000research.134121.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 02/13/2025] Open
Abstract
The haemorrhagic features of viral haemorrhagic fevers may be caused by common patterns of metabolic disturbances of the glucose and ascorbate homeostasis. Haemorrhages and vasculature disfunctions are a clinical feature not only of viral haemorrhagic fevers, but also in scurvy, diabetes and thrombotic microangiopathic haemolytic anaemia. Interestingly, the expression of glucose and ascorbate transporter Glut-1 on the erythrocyte membrane is associated with the inability to synthesize ascorbate and is restricted to that very species that are susceptible to filoviruses (primates, humans and fruit bats). Glut-1 may play a pivotal role in haemorrhagic fever pathogenesis. TIM-1 and TAM receptors have been recognized to enhance entry of Ebola, Lassa and Dengue viruses and viral interferences with TIM-1 could disturb its function, disturbing the expression of Glut-1. In those species not able to synthesize ascorbate and expressing Glut-1 on erythrocytes virus could interact with Glut-1 or other functionally related protein, and the influx of glucose into the cells would be severely impaired. As a consequence, transient hyperglycemia and a marked oxidative stress coupled with the high levels of glucose in plasma would be established, and then promote the activation of NF-κB transcription, exacerbating a pro-inflammatory response mediated by cytokines and chemokines: The inability to synthesize ascorbate is an Achilles Heel when trying to counteract the oxidative stress.
Collapse
Affiliation(s)
- Ivan Chicano Wust
- Universidad Nacional de Educacion a Distancia, Madrid, Community of Madrid, Spain
| |
Collapse
|
5
|
Ma D, Xie A, Lv J, Min X, Zhang X, Zhou Q, Gao D, Wang E, Gao L, Cheng L, Liu S. Engineered extracellular vesicles enable high-efficient delivery of intracellular therapeutic proteins. Protein Cell 2024; 15:724-743. [PMID: 38518087 PMCID: PMC11443452 DOI: 10.1093/procel/pwae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 02/19/2024] [Indexed: 03/24/2024] Open
Abstract
Developing an intracellular delivery system is of key importance in the expansion of protein-based therapeutics acting on cytosolic or nuclear targets. Recently, extracellular vesicles (EVs) have been exploited as next-generation delivery modalities due to their natural role in intercellular communication and biocompatibility. However, fusion of protein of interest to a scaffold represents a widely used strategy for cargo enrichment in EVs, which could compromise the stability and functionality of cargo. Herein, we report intracellular delivery via EV-based approach (IDEA) that efficiently packages and delivers native proteins both in vitro and in vivo without the use of a scaffold. As a proof-of-concept, we applied the IDEA to deliver cyclic GMP-AMP synthase (cGAS), an innate immune sensor. The results showed that cGAS-carrying EVs activated interferon signaling and elicited enhanced antitumor immunity in multiple syngeneic tumor models. Combining cGAS EVs with immune checkpoint inhibition further synergistically boosted antitumor efficacy in vivo. Mechanistically, scRNA-seq demonstrated that cGAS EVs mediated significant remodeling of intratumoral microenvironment, revealing a pivotal role of infiltrating neutrophils in the antitumor immune milieu. Collectively, IDEA, as a universal and facile strategy, can be applied to expand and advance the development of protein-based therapeutics.
Collapse
Affiliation(s)
- Ding Ma
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei 230036, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - An Xie
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei 230036, China
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Jiahui Lv
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Xiaolin Min
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Xinye Zhang
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Qian Zhou
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Daxing Gao
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Enyu Wang
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei 230036, China
| | - Lei Gao
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei 230036, China
| | - Linzhao Cheng
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei 230036, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Senquan Liu
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei 230036, China
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
6
|
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
|
7
|
Roa-Linares VC, Escudero-Flórez M, Vicente-Manzanares M, Gallego-Gómez JC. Host Cell Targets for Unconventional Antivirals against RNA Viruses. Viruses 2023; 15:v15030776. [PMID: 36992484 PMCID: PMC10058429 DOI: 10.3390/v15030776] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/12/2023] [Accepted: 02/28/2023] [Indexed: 03/31/2023] Open
Abstract
The recent COVID-19 crisis has highlighted the importance of RNA-based viruses. The most prominent members of this group are SARS-CoV-2 (coronavirus), HIV (human immunodeficiency virus), EBOV (Ebola virus), DENV (dengue virus), HCV (hepatitis C virus), ZIKV (Zika virus), CHIKV (chikungunya virus), and influenza A virus. With the exception of retroviruses which produce reverse transcriptase, the majority of RNA viruses encode RNA-dependent RNA polymerases which do not include molecular proofreading tools, underlying the high mutation capacity of these viruses as they multiply in the host cells. Together with their ability to manipulate the immune system of the host in different ways, their high mutation frequency poses a challenge to develop effective and durable vaccination and/or treatments. Consequently, the use of antiviral targeting agents, while an important part of the therapeutic strategy against infection, may lead to the selection of drug-resistant variants. The crucial role of the host cell replicative and processing machinery is essential for the replicative cycle of the viruses and has driven attention to the potential use of drugs directed to the host machinery as therapeutic alternatives to treat viral infections. In this review, we discuss small molecules with antiviral effects that target cellular factors in different steps of the infectious cycle of many RNA viruses. We emphasize the repurposing of FDA-approved drugs with broad-spectrum antiviral activity. Finally, we postulate that the ferruginol analog (18-(phthalimide-2-yl) ferruginol) is a potential host-targeted antiviral.
Collapse
Affiliation(s)
- Vicky C Roa-Linares
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| | - Manuela Escudero-Flórez
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, 37007 Salamanca, Spain
| | - Juan C Gallego-Gómez
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| |
Collapse
|
8
|
Pseudotyped Viruses for Marburgvirus and Ebolavirus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1407:105-132. [PMID: 36920694 DOI: 10.1007/978-981-99-0113-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Marburg virus (MARV) and Ebola virus (EBOV) of the Filoviridae family are the most lethal viruses in terms of mortality rate. However, the development of antiviral treatment is hampered by the requirement for biosafety level-4 (BSL-4) containment. The establishment of BSL-2 pseudotyped viruses can provide important tools for the study of filoviruses. This chapter summarizes general information on the filoviruses and then focuses on the construction of replication-deficient pseudotyped MARV and EBOV (e.g., lentivirus system and vesicular stomatitis virus system). It also details the potential applications of the pseudotyped viruses, including neutralization antibody detection, the study of infection mechanisms, the evaluation of antibody-dependent enhancement, virus entry inhibitor screening, and glycoprotein mutation analysis.
Collapse
|
9
|
Solomon M, Liang C. Pseudotyped Viruses for Retroviruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1407:61-84. [PMID: 36920692 DOI: 10.1007/978-981-99-0113-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Since the discovery of retroviruses, their genome and replication strategies have been extensively studied, leading to the discovery of several unique features that make them invaluable vectors for virus pseudotyping, gene delivery, and gene therapy. Notably, retroviral vectors enable the integration of a gene of interest into the host genome, they can be used to stably transduce both dividing and nondividing cells, and they can deliver relatively large genes. Today, retroviral vectors are commonly used for many research applications and have become an active tool in gene therapy and clinical trials. This chapter will discuss the important features of the retroviral genome and replication cycle that are crucial for the development of retroviral vectors, the different retrovirus-based vector systems that are commonly used, and finally the research and clinical applications of retroviral vectors.
Collapse
Affiliation(s)
- Magan Solomon
- Lady Davis Institute, Jewish General Hospital, McGill Centre for Viral Diseases, Montreal, QC, Canada.,Department of Medicine, McGill University, Montreal, QC, Canada
| | - Chen Liang
- Lady Davis Institute, Jewish General Hospital, McGill Centre for Viral Diseases, Montreal, QC, Canada. .,Department of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
10
|
Salampe M, Mamada SS, Evary YM, Mitra S, Bin Emran T, Harapan H, Nainu F, Simal-Gandara J. Promising Marine Natural Products for Tackling Viral Outbreaks: A Focus on Possible Targets and Structure-activity Relationship. Curr Top Med Chem 2023; 23:1352-1379. [PMID: 36045529 DOI: 10.2174/1568026622666220831114838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/11/2022] [Accepted: 05/26/2022] [Indexed: 11/22/2022]
Abstract
Recently, people worldwide have experienced several outbreaks caused by viruses that have attracted much interest globally, such as HIV, Zika, Ebola, and the one being faced, SARSCoV- 2 viruses. Unfortunately, the availability of drugs giving satisfying outcomes in curing those diseases is limited. Therefore, it is necessary to dig deeper to provide compounds that can tackle the causative viruses. Meanwhile, the efforts to explore marine natural products have been gaining great interest as the products have consistently shown several promising biological activities, including antiviral activity. This review summarizes some products extracted from marine organisms, such as seaweeds, seagrasses, sponges, and marine bacteria, reported in recent years to have potential antiviral activities tested through several methods. The mechanisms by which those compounds exert their antiviral effects are also described here, with several main mechanisms closely associated with the ability of the products to block the entry of the viruses into the host cells, inhibiting replication or transcription of the viral genetic material, and disturbing the assembly of viral components. In addition, the structure-activity relationship of the compounds is also highlighted by focusing on six groups of marine compounds, namely sulfated polysaccharides, phlorotannins, terpenoids, lectins, alkaloids, and flavonoids. In conclusion, due to their uniqueness compared to substances extracted from terrestrial sources, marine organisms provide abundant products having promising activities as antiviral agents that can be explored to tackle virus-caused outbreaks.
Collapse
Affiliation(s)
| | - Sukamto Salang Mamada
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, 90245, Indonesia
| | - Yayu Mulsiani Evary
- Department of Pharmaceutical Science and Technology, Faculty of Pharmacy, Hasanuddin University, Makassar, 90245, Indonesia
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka ,1207, Bangladesh
| | - Harapan Harapan
- Department of Microbiology, School of Medicine, Universitas Syiah Kuala, Banda Aceh, 23111, Indonesia
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, 90245, Indonesia
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E32004 Ourense, Spain
| |
Collapse
|
11
|
Wang LL, Estrada L, Wiggins J, Anantpadma M, Patten JJ, Davey RA, Xiang SH. Ligand-based design of peptide entry inhibitors targeting the endosomal receptor binding site of filoviruses. Antiviral Res 2022; 206:105399. [PMID: 36007601 DOI: 10.1016/j.antiviral.2022.105399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 11/25/2022]
Abstract
Filoviruses enter cells through micropinocytosis and trafficking into the endosomes in which they bind to the receptor Niemann-Pick C1 protein (NPC1) for membrane fusion and entry into the cytoplasm. The endosomal receptor-binding is critical step for filovirus entry. Designing inhibitors to block receptor binding will prevent viral entry. Using available binding structural information from the co-crystal structures of the viral GP with the receptor NPC1 or with monoclonal antibodies, we have conducted structure-based design of peptide inhibitors to target the receptor binding site (RBS). The designed peptides were tested for their inhibition activity against pseudo-typed or replication-competent viruses in a cell-based assay. The results indicate that these peptides exhibited strong activities against both Ebola and Marburg virus infection. It is expected that these peptides can be further developed for therapeutic use to treat filovirus infection and combat the outbreaks.
Collapse
Affiliation(s)
- Leah Liu Wang
- School of Veterinary Medicine and Biomedical Sciences, USA; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Leslie Estrada
- School of Veterinary Medicine and Biomedical Sciences, USA; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Joshua Wiggins
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA; School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Manu Anantpadma
- Department of Microbiology & National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, MA, 02115, USA
| | - J J Patten
- Department of Microbiology & National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, MA, 02115, USA
| | - Robert A Davey
- Department of Microbiology & National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, MA, 02115, USA
| | - Shi-Hua Xiang
- School of Veterinary Medicine and Biomedical Sciences, USA; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA.
| |
Collapse
|
12
|
Somiya M, Kuroda S. Reporter gene assay for membrane fusion of extracellular vesicles. J Extracell Vesicles 2021; 10:e12171. [PMID: 34807503 PMCID: PMC8607979 DOI: 10.1002/jev2.12171] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 11/01/2021] [Accepted: 11/08/2021] [Indexed: 01/05/2023] Open
Abstract
Extracellular vesicles (EVs) secreted by living cells are expected to deliver biological cargo molecules, including RNA and proteins, to the cytoplasm of recipient cells. There is an increasing need to understand the mechanism of intercellular cargo delivery by EVs. However, the lack of a feasible bioassay has hampered our understanding of the biological processes of EV uptake, membrane fusion, and cargo delivery to recipient cells. Here, we describe a reporter gene assay that can measure the membrane fusion efficiency of EVs during cargo delivery to recipient cells. When EVs containing tetracycline transactivator (tTA)-fused tetraspanins are internalized by recipient cells and fuse with cell membranes, the tTA domain is exposed to the cytoplasm and cleaved by tobacco etch virus protease to induce tetracycline responsive element (TRE)-mediated reporter gene expression in recipient cells. This assay (designated as EV-mediated tetraspanin-tTA delivery assay, ETTD assay), enabled us to assess the cytoplasmic cargo delivery efficiency of EVs in recipient cells. With the help of a vesicular stomatitis virus-derived membrane fusion protein, the ETTD assay could detect significant enhancement of cargo delivery efficiency of EVs. Furthermore, the ETTD assay could evaluate the effect of potential cargo delivery enhancers/inhibitors. Thus, the ETTD assay may contribute to a better understanding of the underlying mechanism of the cytoplasmic cargo delivery by EVs.
Collapse
Affiliation(s)
- Masaharu Somiya
- SANKEN (The Institute of Scientific and Industrial Research)Osaka UniversityOsakaJapan
| | - Shun'ichi Kuroda
- SANKEN (The Institute of Scientific and Industrial Research)Osaka UniversityOsakaJapan
| |
Collapse
|
13
|
Bhattacharyya S. Mechanisms of Immune Evasion by Ebola Virus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:15-22. [PMID: 34661889 DOI: 10.1007/978-3-030-67452-6_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
The 2013-2016 Ebola virus epidemic in West Africa, which also spread to the USA, UK and Europe, was the largest reported outbreak till date (World Health Organization. 2016. https://apps.who.int/iris/bitstream/handle/10665/208883/ebolasitrep_10Jun2016_eng.pdf;jsessionid=8B7D74BC9D82D2BE1B110BAFFAD3A6E6?sequence=1 ). The recent Ebola outbreak in the Democratic Republic of the Congo has raised immense global concern on this severe and often fatal infection. Although sporadic, the severity and lethality of Ebola virus disease outbreaks has led to extensive research worldwide on this virus. Vaccine (World Health Organization. 2016. https://www.who.int/en/news-room/detail/23-12-2016-final-trial-results-confirm-ebola-vaccine-provides-high-protection-against-disease ; Henao-Restrepo et al. Lancet 389:505-518, 2017) and drug (Hayden. Nature, 557, 475-476, 2018; Dyall et al. J Infect Dis 218(suppl_5), S672-S678, 2018) development efforts against Ebola virus are research hotspots, and a few approved therapeutics are currently available (Centers for Disease Control and Prevention. 2021. https://www.cdc.gov/vhf/ebola/clinicians/vaccine/index.html; Centers for Disease Control and Prevention. 2021. https://www.cdc.gov/vhf/ebola/treatment/index.html). Ebola virus has evolved several mechanisms of host immune evasion, which facilitate its replication and pathogenesis. This chapter describes the Ebola virus morphology, genome, entry, replication, pathogenesis and viral proteins involved in host immune evasion. Further understanding of the underlying molecular mechanisms of immune evasion may facilitate development of additional novel and sustainable strategies against this deadly virus.
Collapse
Affiliation(s)
- Suchita Bhattacharyya
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK.
| |
Collapse
|
14
|
Yang P, Yang Y, Wu Y, Huang C, Ding Y, Wang X, Wang S. An optimized and robust SARS-CoV-2 pseudovirus system for viral entry research. J Virol Methods 2021; 295:114221. [PMID: 34182038 PMCID: PMC8233049 DOI: 10.1016/j.jviromet.2021.114221] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/01/2021] [Accepted: 06/23/2021] [Indexed: 01/08/2023]
Abstract
SARS-CoV-2 is the culprit causing Coronavirus Disease 2019 (COVID-19). For the study of SARS-CoV-2 infection in a BSL-2 laboratory, a SARS-CoV-2 pseudovirus particle (SARS2pp) production and infection system was constructed by using a lentiviral vector bearing dual-reporter genes eGFP and firefly luciferase (Luc2) for easy observation and analysis. Comparison of SARS2pp different production conditions revealed that the pseudovirus titer could be greatly improved by: 1) removing the last 19 amino acids of the spike protein and replacing the signal peptide with the mouse Igk signal sequence; 2) expressing the spike protein using CMV promoter other than CAG (a hybrid promoter consisting of a CMV enhancer, beta-actin promoter, splice donor, and a beta-globin splice acceptor); 3) screening better optimized spike protein sequences for SARS2pp production; and 4) adding 1 % BSA in the SARS2pp production medium. For infection, this SARS2pp system showed a good linear relationship between MOI 2-0.0002 and then was successfully used to evaluate SARS-CoV-2 infection inhibitors including recombinant human ACE2 proteins and SARS-CoV-2 neutralizing antibodies. The kidney, liver and small intestine-derived cell lines were also found to show different susceptibility to SARSpp and SARS2pp. Given its robustness and good performance, it is believed that this pseudovirus particle production and infection system will greatly promote future research for SARS-CoV-2 entry mechanisms and inhibitors and can be easily applied to study new emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Peng Yang
- College of Life Sciences, Henan Normal University, Xinxiang 453007, China; Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yang Yang
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yuming Wu
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Cong Huang
- Beijing Institute of Radiation Medicine, Beijing 100850, China; Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Yanlei Ding
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xuejun Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Shengqi Wang
- College of Life Sciences, Henan Normal University, Xinxiang 453007, China; Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
15
|
Wang LL, Palermo N, Estrada L, Thompson C, Patten JJ, Anantpadma M, Davey RA, Xiang SH. Identification of filovirus entry inhibitors targeting the endosomal receptor NPC1 binding site. Antiviral Res 2021; 189:105059. [PMID: 33705865 PMCID: PMC8088776 DOI: 10.1016/j.antiviral.2021.105059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/27/2021] [Accepted: 03/04/2021] [Indexed: 12/20/2022]
Abstract
Filoviruses, mainly consisting of Ebola viruses (EBOV) and Marburg viruses (MARV), are enveloped negative-strand RNA viruses which can infect humans to cause severe hemorrhagic fevers and outbreaks with high mortality rates. The filovirus infection is mediated by the interaction of viral envelope glycoprotein (GP) and the human endosomal receptor Niemann-Pick C1 (NPC1). Blocking this interaction will prevent the infection. Therefore, we utilized an In silico screening approach to conduct virtual compound screening against the NPC1 receptor-binding site (RBS). Twenty-six top-hit compounds were purchased and evaluated by in vitro cell based inhibition assays against pseudotyped or replication-competent filoviruses. Two classes (A and U) of compounds were identified to have potent inhibitory activity against both Ebola and Marburg viruses. The IC50 values are in the lower level of micromolar concentrations. One compound (compd-A) was found to have a sub-micromolar IC50 value (0.86 μM) against pseudotyped Marburg virus. The cytotoxicity assay (MTT) indicates that compd-A has a moderate cytotoxicity level but the compd-U has much less toxicity and the CC50 value was about 100 μM. Structure-activity relationship (SAR) study has found some analogs of compd-A and -U have reduced the toxicity and enhanced the inhibitory activity. In conclusion, this work has identified several qualified lead-compounds for further drug development against filovirus infection.
Collapse
Affiliation(s)
- Leah Liu Wang
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Nicholas Palermo
- Computational Chemistry Core Facility, VCR Cores, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Leslie Estrada
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Colton Thompson
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - J J Patten
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 0211, USA
| | - Manu Anantpadma
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 0211, USA
| | - Robert A Davey
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 0211, USA
| | - Shi-Hua Xiang
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA.
| |
Collapse
|
16
|
Somiya M, Kuroda S. Real-Time Luminescence Assay for Cytoplasmic Cargo Delivery of Extracellular Vesicles. Anal Chem 2021; 93:5612-5620. [PMID: 33759512 DOI: 10.1021/acs.analchem.1c00339] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Extracellular vesicles (EVs) have been considered to deliver biological cargos between cells and mediate intercellular communication and potential drug delivery carriers. However, the mechanisms that underlie the biological process of EV uptake and cytoplasmic cargo release in recipient cells are largely unknown. Quantitative and real-time assays for the assessment of cargo delivery efficiency inside recipient cells have not been feasible. In this study, we developed an EV cargo delivery (EVCD) assay using a split luciferase called a NanoBiT system. Recipient cells expressing LgBiT, a large subunit of luciferase, emit luminescence when EV cargo proteins fused with a small luminescence tag (HiBiT tag) that can complement LgBiT are delivered to the cytoplasm of recipient cells. Using the EVCD assay, the cargo delivery efficiency of EVs could be quantitatively measured in real time. This assay was highly sensitive in detecting a single event of cargo delivery per cell. We found that modification of EVs with a virus-derived fusogenic protein significantly enhanced the cytoplasmic cargo delivery; however, in the absence of a fusogenic protein, the cargo delivery efficiency of EVs was below the threshold of the assay. The EVCD assay could assess the effect of entry inhibitors on EV cargo delivery. Furthermore, using a luminescence microscope, the cytoplasmic cargo delivery of EVs was directly visualized in living cells. This assay could reveal the biological mechanism of the cargo delivery processes of EVs.
Collapse
Affiliation(s)
- Masaharu Somiya
- Department of Biomolecular Science and Reaction, The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan
| | - Shun'ichi Kuroda
- Department of Biomolecular Science and Reaction, The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan
| |
Collapse
|
17
|
Lee J, Kreutzberger AJB, Odongo L, Nelson EA, Nyenhuis DA, Kiessling V, Liang B, Cafiso DS, White JM, Tamm LK. Ebola virus glycoprotein interacts with cholesterol to enhance membrane fusion and cell entry. Nat Struct Mol Biol 2021; 28:181-189. [PMID: 33462517 PMCID: PMC7992113 DOI: 10.1038/s41594-020-00548-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 12/07/2020] [Indexed: 12/14/2022]
Abstract
Cholesterol serves critical roles in enveloped virus fusion by modulating membrane properties. The glycoprotein (GP) of Ebola virus (EBOV) promotes fusion in the endosome, a process that requires the endosomal cholesterol transporter NPC1. However, the role of cholesterol in EBOV fusion is unclear. Here we show that cholesterol in GP-containing membranes enhances fusion and the membrane-proximal external region and transmembrane (MPER/TM) domain of GP interacts with cholesterol via several glycine residues in the GP2 TM domain, notably G660. Compared to wild-type (WT) counterparts, a G660L mutation caused a more open angle between MPER and TM domains in an MPER/TM construct, higher probability of stalling at hemifusion for GP2 proteoliposomes and lower cell entry of virus-like particles (VLPs). VLPs with depleted cholesterol show reduced cell entry, and VLPs produced under cholesterol-lowering statin conditions show less frequent entry than respective controls. We propose that cholesterol-TM interactions affect structural features of GP2, thereby facilitating fusion and cell entry.
Collapse
Affiliation(s)
- Jinwoo Lee
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, USA
| | - Alex J B Kreutzberger
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Laura Odongo
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Elizabeth A Nelson
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA
| | - David A Nyenhuis
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
- Department of Chemistry, University of Virginia, Charlottesville, VA, USA
| | - Volker Kiessling
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Binyong Liang
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - David S Cafiso
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
- Department of Chemistry, University of Virginia, Charlottesville, VA, USA
| | - Judith M White
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA
| | - Lukas K Tamm
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA.
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
18
|
Christy MP, Uekusa Y, Gerwick L, Gerwick WH. Natural Products with Potential to Treat RNA Virus Pathogens Including SARS-CoV-2. JOURNAL OF NATURAL PRODUCTS 2021; 84:161-182. [PMID: 33352046 PMCID: PMC7771248 DOI: 10.1021/acs.jnatprod.0c00968] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Indexed: 05/03/2023]
Abstract
Three families of RNA viruses, the Coronaviridae, Flaviviridae, and Filoviridae, collectively have great potential to cause epidemic disease in human populations. The current SARS-CoV-2 (Coronaviridae) responsible for the COVID-19 pandemic underscores the lack of effective medications currently available to treat these classes of viral pathogens. Similarly, the Flaviviridae, which includes such viruses as Dengue, West Nile, and Zika, and the Filoviridae, with the Ebola-type viruses, as examples, all lack effective therapeutics. In this review, we present fundamental information concerning the biology of these three virus families, including their genomic makeup, mode of infection of human cells, and key proteins that may offer targeted therapies. Further, we present the natural products and their derivatives that have documented activities to these viral and host proteins, offering hope for future mechanism-based antiviral therapeutics. By arranging these potential protein targets and their natural product inhibitors by target type across these three families of virus, new insights are developed, and crossover treatment strategies are suggested. Hence, natural products, as is the case for other therapeutic areas, continue to be a promising source of structurally diverse new anti-RNA virus therapeutics.
Collapse
Affiliation(s)
- Mitchell P. Christy
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California 92093, United States
| | - Yoshinori Uekusa
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California 92093, United States
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Lena Gerwick
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California 92093, United States
| | - William H. Gerwick
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California 92093, United States
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
19
|
Hoornweg TE, Bouma EM, van de Pol DP, Rodenhuis-Zybert IA, Smit JM. Chikungunya virus requires an intact microtubule network for efficient viral genome delivery. PLoS Negl Trop Dis 2020; 14:e0008469. [PMID: 32764759 PMCID: PMC7413472 DOI: 10.1371/journal.pntd.0008469] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/10/2020] [Indexed: 11/25/2022] Open
Abstract
Chikungunya virus (CHIKV) is a re-emerging mosquito-borne alphavirus, which has rapidly spread around the globe thereby causing millions of infections. CHIKV is an enveloped virus belonging to the Togaviridae family and enters its host cell primarily via clathrin-mediated endocytosis. Upon internalization, the endocytic vesicle containing the virus particle moves through the cell and delivers the virus to early endosomes where membrane fusion is observed. Thereafter, the nucleocapsid dissociates and the viral RNA is translated into proteins. In this study, we examined the importance of the microtubule network during the early steps of infection and dissected the intracellular trafficking behavior of CHIKV particles during cell entry. We observed two distinct CHIKV intracellular trafficking patterns prior to membrane hemifusion. Whereas half of the CHIKV virions remained static during cell entry and fused in the cell periphery, the other half showed fast-directed microtubule-dependent movement prior to delivery to Rab5-positive early endosomes and predominantly fused in the perinuclear region of the cell. Disruption of the microtubule network reduced the number of infected cells. At these conditions, membrane hemifusion activity was not affected yet fusion was restricted to the cell periphery. Furthermore, follow-up experiments revealed that disruption of the microtubule network impairs the delivery of the viral genome to the cell cytosol. We therefore hypothesize that microtubules may direct the particle to a cellular location that is beneficial for establishing infection or aids in nucleocapsid uncoating. Chikungunya virus (CHIKV) is an alphavirus that is transmitted to humans by infected mosquitoes. Disease symptoms can include fever, rash, myalgia, and long-lasting debilitating joint pains. Unfortunately, there is currently no licensed vaccine or antiviral treatment available to combat CHIKV. Understanding the virus:host interactions during the replication cycle of the virus is crucial for the development of effective antiviral therapies. In this study we elucidated the trafficking behavior of CHIKV particles early in infection. During cell entry, CHIKV virions require an intact microtubule network for efficient delivery of the viral genome into the host cell thereby increasing the chance to productively infect a cell.
Collapse
Affiliation(s)
- Tabitha E. Hoornweg
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Ellen M. Bouma
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Denise P.I. van de Pol
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Izabela A. Rodenhuis-Zybert
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Jolanda M. Smit
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, The Netherlands
- * E-mail:
| |
Collapse
|
20
|
Efficient Expression and Processing of Ebola Virus Glycoprotein Induces Morphological Changes in BmN Cells but Cannot Rescue Deficiency of Bombyx Mori Nucleopolyhedrovirus GP64. Viruses 2019; 11:v11111067. [PMID: 31731691 PMCID: PMC6893839 DOI: 10.3390/v11111067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 01/08/2023] Open
Abstract
Ebola virus (EBOV) disease outbreaks have resulted in many fatalities, yet no licensed vaccines are available to prevent infection. Recombinant glycoprotein (GP) production may contribute to finding a cure for Ebola virus disease, which is the key candidate protein for vaccine preparation. To explore GP1,2 expression in BmN cells, EBOV-GP1,2 with its native signal peptide or the GP64 signal peptide was cloned and transferred into a normal or gp64 null Bombyx mori nucleopolyhedrovirus (BmNPV) bacmid via transposition. The infectivity of the recombinant bacmids was investigated after transfection, expression and localization of EBOV-GP were investigated, and cell morphological changes were analyzed by TEM. The GP64 signal peptide, but not the GP1,2 native signal peptide, caused GP1,2 localization to the cell membrane, and the differentially localized GP1,2 proteins were cleaved into GP1 and GP2 fragments in BmN cells. GP1,2 expression resulted in dramatic morphological changes in BmN cells in the early stage of infection. However, GP1,2 expression did not rescue GP64 deficiency in BmNPV infection. This study provides a better understanding of GP expression and processing in BmN cells, which may lay a foundation for EBOV-GP expression using the BmNPV baculovirus expression system.
Collapse
|
21
|
Mirza MU, Vanmeert M, Ali A, Iman K, Froeyen M, Idrees M. Perspectives towards antiviral drug discovery against Ebola virus. J Med Virol 2019; 91:2029-2048. [PMID: 30431654 PMCID: PMC7166701 DOI: 10.1002/jmv.25357] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/04/2018] [Indexed: 12/18/2022]
Abstract
Ebola virus disease (EVD), caused by Ebola viruses, resulted in more than 11 500 deaths according to a recent 2018 WHO report. With mortality rates up to 90%, it is nowadays one of the most deadly infectious diseases. However, no Food and Drug Administration‐approved Ebola drugs or vaccines are available yet with the mainstay of therapy being supportive care. The high fatality rate and absence of effective treatment or vaccination make Ebola virus a category‐A biothreat pathogen. Fortunately, a series of investigational countermeasures have been developed to control and prevent this global threat. This review summarizes the recent therapeutic advances and ongoing research progress from research and development to clinical trials in the development of small‐molecule antiviral drugs, small‐interference RNA molecules, phosphorodiamidate morpholino oligomers, full‐length monoclonal antibodies, and vaccines. Moreover, difficulties are highlighted in the search for effective countermeasures against EVD with additional focus on the interplay between available in silico prediction methods and their evidenced potential in antiviral drug discovery.
Collapse
Affiliation(s)
- Muhammad Usman Mirza
- Department of Pharmaceutical Sciences, REGA Institute for Medical Research, Medicinal Chemistry, KU Leuven, Leuven, Belgium
| | - Michiel Vanmeert
- Department of Pharmaceutical Sciences, REGA Institute for Medical Research, Medicinal Chemistry, KU Leuven, Leuven, Belgium
| | - Amjad Ali
- Department of Genetics, Hazara University, Mansehra, Pakistan.,Molecular Virology Laboratory, Centre for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Kanzal Iman
- Biomedical Informatics Research Laboratory (BIRL), Department of Biology, Lahore University of Management Sciences (LUMS), Lahore, Pakistan
| | - Matheus Froeyen
- Department of Pharmaceutical Sciences, REGA Institute for Medical Research, Medicinal Chemistry, KU Leuven, Leuven, Belgium
| | - Muhammad Idrees
- Molecular Virology Laboratory, Centre for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan.,Hazara University Mansehra, Khyber Pakhtunkhwa Pakistan
| |
Collapse
|
22
|
Olejnik J, Hume AJ, Leung DW, Amarasinghe GK, Basler CF, Mühlberger E. Filovirus Strategies to Escape Antiviral Responses. Curr Top Microbiol Immunol 2019; 411:293-322. [PMID: 28685291 PMCID: PMC5973841 DOI: 10.1007/82_2017_13] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This chapter describes the various strategies filoviruses use to escape host immune responses with a focus on innate immune and cell death pathways. Since filovirus replication can be efficiently blocked by interferon (IFN), filoviruses have evolved mechanisms to counteract both type I IFN induction and IFN response signaling pathways. Intriguingly, marburg- and ebolaviruses use different strategies to inhibit IFN signaling. This chapter also summarizes what is known about the role of IFN-stimulated genes (ISGs) in filovirus infection. These fall into three categories: those that restrict filovirus replication, those whose activation is inhibited by filoviruses, and those that have no measurable effect on viral replication. In addition to innate immunity, mammalian cells have evolved strategies to counter viral infections, including the induction of cell death and stress response pathways, and we summarize our current knowledge of how filoviruses interact with these pathways. Finally, this chapter delves into the interaction of EBOV with myeloid dendritic cells and macrophages and the associated inflammatory response, which differs dramatically between these cell types when they are infected with EBOV. In summary, we highlight the multifaceted nature of the host-viral interactions during filoviral infections.
Collapse
Affiliation(s)
- Judith Olejnik
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, Boston, MA, 02118, USA
| | - Adam J Hume
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, Boston, MA, 02118, USA
| | - Daisy W Leung
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Christopher F Basler
- Microbial Pathogenesis, Georgia State University, Institute for Biomedical Sciences, Atlanta, GA, 30303, USA
| | - Elke Mühlberger
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, Boston, MA, 02118, USA.
| |
Collapse
|
23
|
Ao Z, Wang L, Mendoza EJ, Cheng K, Zhu W, Cohen EA, Fowke K, Qiu X, Kobinger G, Yao X. Incorporation of Ebola glycoprotein into HIV particles facilitates dendritic cell and macrophage targeting and enhances HIV-specific immune responses. PLoS One 2019; 14:e0216949. [PMID: 31100082 PMCID: PMC6524799 DOI: 10.1371/journal.pone.0216949] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 05/01/2019] [Indexed: 01/05/2023] Open
Abstract
The development of an effective vaccine against HIV infection remains a global priority. Dendritic cell (DC)-based HIV immunotherapeutic vaccine is a promising approach which aims at optimizing the HIV-specific immune response using primed DCs to promote and enhance both the cellular and humoral arms of immunity. Since the Ebola virus envelope glycoprotein (EboGP) has strong DC-targeting ability, we investigated whether EboGP is able to direct HIV particles towards DCs efficiently and promote potent HIV-specific immune responses. Our results indicate that the incorporation of EboGP into non-replicating virus-like particles (VLPs) enhances their ability to target human monocyte-derived dendritic cells (MDDCs) and monocyte-derived macrophages (MDMs). Also, a mucin-like domain deleted EboGP (EboGPΔM) can further enhanced the MDDCs and MDMs-targeting ability. Furthermore, we investigated the effect of EboGP on HIV immunogenicity in mice, and the results revealed a significantly stronger HIV-specific humoral immune response when immunized with EboGP-pseudotyped HIV VLPs compared with those immunized with HIV VLPs. Splenocytes harvested from mice immunized with EboGP-pseudotyped HIV VLPs secreted increased levels of macrophage inflammatory proteins-1α (MIP-1α) and IL-4 upon stimulation with HIV Env and/or Gag peptides compared with those harvested from mice immunized with HIV VLPs. Collectively, this study provides evidence for the first time that the incorporation of EboGP in HIV VLPs can facilitate DC and macrophage targeting and induce more potent immune responses against HIV.
Collapse
MESH Headings
- AIDS Vaccines/administration & dosage
- AIDS Vaccines/genetics
- AIDS Vaccines/immunology
- Animals
- Chemokine CCL3/genetics
- Chemokine CCL3/immunology
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/virology
- Ebolavirus/chemistry
- Female
- Gene Expression
- HEK293 Cells
- HIV Antibodies/biosynthesis
- HIV Infections/immunology
- HIV Infections/prevention & control
- HIV Infections/virology
- HIV-1/drug effects
- HIV-1/growth & development
- HIV-1/immunology
- Humans
- Immunity, Cellular/drug effects
- Immunity, Humoral/drug effects
- Immunization
- Immunogenicity, Vaccine
- Interleukin-4/genetics
- Interleukin-4/immunology
- Lymphocytes/cytology
- Lymphocytes/drug effects
- Lymphocytes/immunology
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/virology
- Mice
- Mice, Inbred C57BL
- Molecular Targeted Therapy
- Primary Cell Culture
- Spleen/cytology
- Spleen/drug effects
- Spleen/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- env Gene Products, Human Immunodeficiency Virus/genetics
- env Gene Products, Human Immunodeficiency Virus/immunology
- gag Gene Products, Human Immunodeficiency Virus/genetics
- gag Gene Products, Human Immunodeficiency Virus/immunology
Collapse
Affiliation(s)
- Zhujun Ao
- Laboratory of Molecular Human Retrovirology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lijun Wang
- Laboratory of Molecular Human Retrovirology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Histology and Embryology, Zunyi Medical College, Zunyi, Guizhou, China
| | - Emelissa J. Mendoza
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Keding Cheng
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Wenjun Zhu
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Eric A. Cohen
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada
| | - Keith Fowke
- Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Xiangguo Qiu
- Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Gary Kobinger
- Centre de Recherche en Infectiologie de l’Université Laval/Centre Hospitalier de l’Université Laval (CHUL), Québec, Quebec, Canada
- * E-mail: (XJY); (GK)
| | - Xiaojian Yao
- Laboratory of Molecular Human Retrovirology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- * E-mail: (XJY); (GK)
| |
Collapse
|
24
|
Xia S, Yan L, Xu W, Agrawal AS, Algaissi A, Tseng CTK, Wang Q, Du L, Tan W, Wilson IA, Jiang S, Yang B, Lu L. A pan-coronavirus fusion inhibitor targeting the HR1 domain of human coronavirus spike. SCIENCE ADVANCES 2019; 5:eaav4580. [PMID: 30989115 PMCID: PMC6457931 DOI: 10.1126/sciadv.aav4580] [Citation(s) in RCA: 363] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 02/14/2019] [Indexed: 05/07/2023]
Abstract
Continuously emerging highly pathogenic human coronaviruses (HCoVs) remain a major threat to human health, as illustrated in past SARS-CoV and MERS-CoV outbreaks. The development of a drug with broad-spectrum HCoV inhibitory activity would address this urgent unmet medical need. Although previous studies have suggested that the HR1 of HCoV spike (S) protein is an important target site for inhibition against specific HCoVs, whether this conserved region could serve as a target for the development of broad-spectrum pan-CoV inhibitor remains controversial. Here, we found that peptide OC43-HR2P, derived from the HR2 domain of HCoV-OC43, exhibited broad fusion inhibitory activity against multiple HCoVs. EK1, the optimized form of OC43-HR2P, showed substantially improved pan-CoV fusion inhibitory activity and pharmaceutical properties. Crystal structures indicated that EK1 can form a stable six-helix bundle structure with both short α-HCoV and long β-HCoV HR1s, further supporting the role of HR1 region as a viable pan-CoV target site.
Collapse
Affiliation(s)
- Shuai Xia
- Shanghai Public Health Clinical Center and School of Basic Medical Sciences, and Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 200032, China
| | - Lei Yan
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Wei Xu
- Shanghai Public Health Clinical Center and School of Basic Medical Sciences, and Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 200032, China
| | - Anurodh Shankar Agrawal
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Abdullah Algaissi
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Chien-Te K. Tseng
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Qian Wang
- Shanghai Public Health Clinical Center and School of Basic Medical Sciences, and Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 200032, China
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
| | - Wenjie Tan
- MOH Key Laboratory of Medical Virology, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ian A. Wilson
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
- Department of Integrative Structural and Computational Biology and the Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, BCC206, La Jolla, CA 92037, USA
- Corresponding author. (I.A.W.); (S.J.); (B.Y.); (L.L.)
| | - Shibo Jiang
- Shanghai Public Health Clinical Center and School of Basic Medical Sciences, and Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 200032, China
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
- Corresponding author. (I.A.W.); (S.J.); (B.Y.); (L.L.)
| | - Bei Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
- Corresponding author. (I.A.W.); (S.J.); (B.Y.); (L.L.)
| | - Lu Lu
- Shanghai Public Health Clinical Center and School of Basic Medical Sciences, and Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 200032, China
- Corresponding author. (I.A.W.); (S.J.); (B.Y.); (L.L.)
| |
Collapse
|
25
|
Abstract
The membrane fusion properties of HCV envelope glycoproteins can be evaluated using several assays. Fusion assays generally require contacts between glycoproteins expressed on a donor membrane, such as those from a cell or a viral particle, and an acceptor membrane that may or may not express cognate viral receptor, such as those from an indicator cell or a liposome. In this chapter, we describe three well-established methods in the field that use either cell surface expression of glycoproteins, HCV pseudoparticles (HCVpp), or cell culture-grown HCV (HCVcc) particles for donor membrane and cells or liposomes as acceptor membrane in which specific components can be included to monitor and quantify fusion. We provide details of cell-cell fusion assay, virus-liposome fusion assay, and finally virus-plasma membrane fusion assay. We also describe inhibitors that can block HCV envelope membrane fusion.
Collapse
|
26
|
Schindell BG, Webb AL, Kindrachuk J. Persistence and Sexual Transmission of Filoviruses. Viruses 2018; 10:E683. [PMID: 30513823 PMCID: PMC6316729 DOI: 10.3390/v10120683] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 12/27/2022] Open
Abstract
There is an increasing frequency of reports regarding the persistence of the Ebola virus (EBOV) in Ebola virus disease (EVD) survivors. During the 2014⁻2016 West African EVD epidemic, sporadic transmission events resulted in the initiation of new chains of human-to-human transmission. Multiple reports strongly suggest that these re-emergences were linked to persistent EBOV infections and included sexual transmission from EVD survivors. Asymptomatic infection and long-term viral persistence in EVD survivors could result in incidental introductions of the Ebola virus in new geographic regions and raise important national and local public health concerns. Alarmingly, although the persistence of filoviruses and their potential for sexual transmission have been documented since the emergence of such viruses in 1967, there is limited knowledge regarding the events that result in filovirus transmission to, and persistence within, the male reproductive tract. Asymptomatic infection and long-term viral persistence in male EVD survivors could lead to incidental transfer of EBOV to new geographic regions, thereby generating widespread outbreaks that constitute a significant threat to national and global public health. Here, we review filovirus testicular persistence and discuss the current state of knowledge regarding the rates of persistence in male survivors, and mechanisms underlying reproductive tract localization and sexual transmission.
Collapse
Affiliation(s)
- Brayden G Schindell
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Andrew L Webb
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Jason Kindrachuk
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| |
Collapse
|
27
|
Fanunza E, Frau A, Corona A, Tramontano E. Antiviral Agents Against Ebola Virus Infection: Repositioning Old Drugs and Finding Novel Small Molecules. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2018; 51:135-173. [PMID: 32287476 PMCID: PMC7112331 DOI: 10.1016/bs.armc.2018.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Ebola virus (EBOV) causes a deadly hemorrhagic syndrome in humans with mortality rate up to 90%. First reported in Zaire in 1976, EBOV outbreaks showed a fluctuating trend during time and fora long period it was considered a tragic disease confined to the isolated regions of the African continent where the EBOV fear was perpetuated among the poor communities. The extreme severity of the recent 2014-16 EBOV outbreak in terms of fatality rate and rapid spread out of Africa led to the understanding that EBOV is a global health risk and highlights the necessity to find countermeasures against it. In the recent years, several small molecules have been shown to display in vitro and in vivo efficacy against EBOV and some of them have advanced into clinical trials. In addition, also existing drugs have been tested for their anti-EBOV activity and were shown to be promising candidates. However, despite the constant effort addressed to identify anti-EBOV therapeutics, no approved drugs are available against EBOV yet. In this chapter, we describe the main EBOV life cycle steps, providing a detailed picture of the druggable viral and host targets that have been explored so far by different technologies. We then summarize the small molecules, nucleic acid oligomers, and antibody-based therapies reported to have an effect either in in silico, or in biochemical and cell-based assays or in animal models and clinical trials, listing them according to their demonstrated or putative mechanism of action.
Collapse
Affiliation(s)
- Elisa Fanunza
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Aldo Frau
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Angela Corona
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
- Genetics and Biomedical Research Institute, National Research Council, Monserrato, Italy
| |
Collapse
|
28
|
Dhama K, Karthik K, Khandia R, Chakraborty S, Munjal A, Latheef SK, Kumar D, Ramakrishnan MA, Malik YS, Singh R, Malik SVS, Singh RK, Chaicumpa W. Advances in Designing and Developing Vaccines, Drugs, and Therapies to Counter Ebola Virus. Front Immunol 2018; 9:1803. [PMID: 30147687 PMCID: PMC6095993 DOI: 10.3389/fimmu.2018.01803] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 07/23/2018] [Indexed: 01/10/2023] Open
Abstract
Ebola virus (EBOV), a member of the family Filoviridae, is responsible for causing Ebola virus disease (EVD) (formerly named Ebola hemorrhagic fever). This is a severe, often fatal illness with mortality rates varying from 50 to 90% in humans. Although the virus and associated disease has been recognized since 1976, it was only when the recent outbreak of EBOV in 2014-2016 highlighted the danger and global impact of this virus, necessitating the need for coming up with the effective vaccines and drugs to counter its pandemic threat. Albeit no commercial vaccine is available so far against EBOV, a few vaccine candidates are under evaluation and clinical trials to assess their prophylactic efficacy. These include recombinant viral vector (recombinant vesicular stomatitis virus vector, chimpanzee adenovirus type 3-vector, and modified vaccinia Ankara virus), Ebola virus-like particles, virus-like replicon particles, DNA, and plant-based vaccines. Due to improvement in the field of genomics and proteomics, epitope-targeted vaccines have gained top priority. Correspondingly, several therapies have also been developed, including immunoglobulins against specific viral structures small cell-penetrating antibody fragments that target intracellular EBOV proteins. Small interfering RNAs and oligomer-mediated inhibition have also been verified for EVD treatment. Other treatment options include viral entry inhibitors, transfusion of convalescent blood/serum, neutralizing antibodies, and gene expression inhibitors. Repurposed drugs, which have proven safety profiles, can be adapted after high-throughput screening for efficacy and potency for EVD treatment. Herbal and other natural products are also being explored for EVD treatment. Further studies to better understand the pathogenesis and antigenic structures of the virus can help in developing an effective vaccine and identifying appropriate antiviral targets. This review presents the recent advances in designing and developing vaccines, drugs, and therapies to counter the EBOV threat.
Collapse
Affiliation(s)
- Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Rekha Khandia
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, India
| | - Sandip Chakraborty
- Department of Veterinary Microbiology, College of Veterinary Sciences and Animal Husbandry, Agartala, India
| | - Ashok Munjal
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, India
| | - Shyma K. Latheef
- Immunology Section, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Deepak Kumar
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | | | - Yashpal Singh Malik
- Division of Biological Standardization, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Rajendra Singh
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Satya Veer Singh Malik
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Raj Kumar Singh
- ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Wanpen Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine SIriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
29
|
Rumlová M, Ruml T. In vitro methods for testing antiviral drugs. Biotechnol Adv 2018; 36:557-576. [PMID: 29292156 PMCID: PMC7127693 DOI: 10.1016/j.biotechadv.2017.12.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/22/2017] [Accepted: 12/27/2017] [Indexed: 12/24/2022]
Abstract
Despite successful vaccination programs and effective treatments for some viral infections, humans are still losing the battle with viruses. Persisting human pandemics, emerging and re-emerging viruses, and evolution of drug-resistant strains impose continuous search for new antiviral drugs. A combination of detailed information about the molecular organization of viruses and progress in molecular biology and computer technologies has enabled rational antivirals design. Initial step in establishing efficacy of new antivirals is based on simple methods assessing inhibition of the intended target. We provide here an overview of biochemical and cell-based assays evaluating the activity of inhibitors of clinically important viruses.
Collapse
Affiliation(s)
- Michaela Rumlová
- Department of Biotechnology, University of Chemistry and Technology, Prague 166 28, Czech Republic.
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague 166 28, Czech Republic.
| |
Collapse
|
30
|
Hulseberg CE, Fénéant L, Szymańska KM, White JM. Lamp1 Increases the Efficiency of Lassa Virus Infection by Promoting Fusion in Less Acidic Endosomal Compartments. mBio 2018; 9:e01818-17. [PMID: 29295909 PMCID: PMC5750398 DOI: 10.1128/mbio.01818-17] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 11/21/2017] [Indexed: 01/20/2023] Open
Abstract
Lassa virus (LASV) is an arenavirus whose entry into host cells is mediated by a glycoprotein complex (GPC) comprised of a receptor binding subunit, GP1, a fusogenic transmembrane subunit, GP2, and a stable signal peptide. After receptor-mediated internalization, arenaviruses converge in the endocytic pathway, where they are thought to undergo low-pH-triggered, GPC-mediated fusion with a late endosome membrane. A unique feature of LASV entry is a pH-dependent switch from a primary cell surface receptor (α-dystroglycan) to an endosomal receptor, lysosomal-associated membrane protein (Lamp1). Despite evidence that the interaction between LASV GP1 and Lamp1 is critical, the function of Lamp1 in promoting LASV infection remains poorly characterized. Here we used wild-type (WT) and Lamp1 knockout (KO) cells to show that Lamp1 increases the efficiency of, but is not absolutely required for, LASV entry and infection. We then used cell-cell and pseudovirus-cell surface fusion assays to demonstrate that LASV GPC-mediated fusion occurs at a significantly higher pH when Lamp1 is present compared to when Lamp1 is missing. Correspondingly, we found that LASV entry occurs through less acidic endosomes in WT (Lamp1-positive) versus Lamp1 KO cells. We propose that, by elevating the pH threshold for fusion, Lamp1 allows LASV particles to exit the endocytic pathway before they encounter an increasingly acidic and harsh proteolytic environment, which could inactivate a significant percentage of incoming viruses. In this manner Lamp1 increases the overall efficiency of LASV entry and infection.IMPORTANCE Lassa virus is the most clinically important member of the Arenaviridae, a family that includes six additional biosafety level 4 (BSL4) hemorrhagic fever viruses. The lack of specific antiviral therapies for Lassa fever drives an urgent need to identify druggable targets, and interventions that block infection at the entry stage are particularly attractive. Lassa virus is only the second virus known to employ an intracellular receptor, the first being Ebola virus. Here we show that interaction with its intracellular receptor, Lamp1, enhances and upwardly shifts the pH dependence of fusion and consistently permits Lassa virus entry into cells through less acidic endosomes. We propose that in this manner, Lamp1 increases the overall efficiency of Lassa virus infection.
Collapse
Affiliation(s)
| | - Lucie Fénéant
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | | | - Judith M White
- Department of Microbiology, University of Virginia, Charlottesville, Virginia, USA
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
31
|
Dehghani B, Ghasabi F, Hashempoor T, Joulaei H, Hasanshahi Z, Halaji M, Chatrabnous N, Mousavi Z, Moayedi J. Functional and structural characterization of Ebola virus glycoprotein (1976–2015) — Anin silicostudy. INT J BIOMATH 2017. [DOI: 10.1142/s179352451750108x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Ebola virus (EBOV) is the causative agent of a severe hemorrhagic fever disease associated with high mortality rates in humans. This virus has five strains of which Zaire Ebola virus (ZEBOV) is the first and most important strain. It can be transmitted through contact with contaminated surfaces and objects. The genome of EBOV codes one non-structural and seven structural proteins consisting of two forms of glycoprotein (GP): soluble glycoprotein (sGP) and GP (spike). In this paper, we attempted to characterize and predict physicochemical properties, B-cell epitopes, mutation sites, phosphorylation sites, glycosylation sites, and different protein structures of EBOV GP to provide comprehensive data about changes of this GP during a 40-years course (1976–2015). GP sequences were obtained from NCBI gene bank from 1976–2015. Expasy’sProtParam, PROTSCALE, immuneepitope, Bepipred, BcePred, ABCpred, VaxiJen, DISPHOS, NetPhos, and numerous programs were used to predict and analyze all sequences. More variety of mutations were found in 2015 sequences and mutations were related to huge changes in B-cell epitopes, phosphorylation and glycosylation sites. In addition, our results determined different sites of disulfide bonds and an important mutation related to IgE epitope as well as four potent B-cell epitopes (380–387, 318–338, 405–438 and 434–475). In this study, we suggested the effect of mutations on GP properties, six positions for disulfide bonds and four phosphorylation sites for protein kinase C enzyme. Selected sequences were shown different sites for O-link and N-link glycosylation. A mutation that changed GP to an allergen protein and has a significant role in vaccine designing as well as four potent B-cell epitopes in GP protein were found.
Collapse
Affiliation(s)
- Behzad Dehghani
- Shiraz HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzane Ghasabi
- Shiraz HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tayebeh Hashempoor
- Shiraz HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hassan Joulaei
- Shiraz HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Hasanshahi
- Shiraz HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrdad Halaji
- Shiraz HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Microbiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, IR Iran
| | - Nazanin Chatrabnous
- Shiraz HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Mousavi
- Shiraz HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Javad Moayedi
- Shiraz HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
32
|
King B, Tarr AW. How have retrovirus pseudotypes contributed to our understanding of viral entry? Future Virol 2017. [DOI: 10.2217/fvl-2017-0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Study of virus entry into host cells is important for understanding viral tropism and pathogenesis. Studying the entry of in vitro cultured viruses is not always practicable. Study of highly pathogenic viruses, viruses that do not grow in culture, and viruses that rapidly change phenotype in vitro can all benefit from alternative models of entry. Retrovirus particles can be engineered to display the envelope proteins of heterologous enveloped viruses. This approach, broadly termed ‘pseudotyping’, is an important technique for interrogating virus entry. In this perspective we consider how retrovirus pseudotypes have addressed these challenges and improved our understanding of the entry pathways of diverse virus species, including Ebolavirus, human immunodeficiency virus and hepatitis C virus.
Collapse
Affiliation(s)
- Barnabas King
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust & the University of Nottingham, Nottingham, UK
- School of Life Sciences, Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham, UK
| | - Alexander W Tarr
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust & the University of Nottingham, Nottingham, UK
- School of Life Sciences, Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
33
|
Structure of the Ebola virus envelope protein MPER/TM domain and its interaction with the fusion loop explains their fusion activity. Proc Natl Acad Sci U S A 2017; 114:E7987-E7996. [PMID: 28874543 DOI: 10.1073/pnas.1708052114] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Ebolavirus (EBOV), an enveloped filamentous RNA virus causing severe hemorrhagic fever, enters cells by macropinocytosis and membrane fusion in a late endosomal compartment. Fusion is mediated by the EBOV envelope glycoprotein GP, which consists of subunits GP1 and GP2. GP1 binds to cellular receptors, including Niemann-Pick C1 (NPC1) protein, and GP2 is responsible for low pH-induced membrane fusion. Proteolytic cleavage and NPC1 binding at endosomal pH lead to conformational rearrangements of GP2 that include exposing the hydrophobic fusion loop (FL) for insertion into the cellular target membrane and forming a six-helix bundle structure. Although major portions of the GP2 structure have been solved in pre- and postfusion states and although current models place the transmembrane (TM) and FL domains of GP2 in close proximity at critical steps of membrane fusion, their structures in membrane environments, and especially interactions between them, have not yet been characterized. Here, we present the structure of the membrane proximal external region (MPER) connected to the TM domain: i.e., the missing parts of the EBOV GP2 structure. The structure, solved by solution NMR and EPR spectroscopy in membrane-mimetic environments, consists of a helix-turn-helix architecture that is independent of pH. Moreover, the MPER region is shown to interact in the membrane interface with the previously determined structure of the EBOV FL through several critical aromatic residues. Mutation of aromatic and neighboring residues in both binding partners decreases fusion and viral entry, highlighting the functional importance of the MPER/TM-FL interaction in EBOV entry and fusion.
Collapse
|
34
|
Selective inhibition of Ebola entry with selective estrogen receptor modulators by disrupting the endolysosomal calcium. Sci Rep 2017; 7:41226. [PMID: 28117364 PMCID: PMC5259750 DOI: 10.1038/srep41226] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 12/19/2016] [Indexed: 12/15/2022] Open
Abstract
The Ebola crisis occurred in West-Africa highlights the urgency for its clinical treatments. Currently, no Food and Drug Administration (FDA)-approved therapeutics are available. Several FDA-approved drugs, including selective estrogen receptor modulators (SERMs), possess selective anti-Ebola activities. However, the inhibitory mechanisms of these drugs remain elusive. By analyzing the structures of SERMs and their incidental biological activity (cholesterol accumulation), we hypothesized that this incidental biological activity induced by SERMs could be a plausible mechanism as to their inhibitory effects on Ebola infection. Herein, we demonstrated that the same dosages of SERMs which induced cholesterol accumulation also inhibited Ebola infection. SERMs reduced the cellular sphingosine and subsequently caused endolysosomal calcium accumulation, which in turn led to blocking the Ebola entry. Our study clarified the specific anti-Ebola mechanism of SERMs, even the cationic amphiphilic drugs (CADs), this mechanism led to the endolysosomal calcium as a critical target for development of anti-Ebola drugs.
Collapse
|
35
|
Holic N, Frin S, Seye AK, Galy A, Fenard D. Improvement of De Novo Cholesterol Biosynthesis Efficiently Promotes the Production of Human Immunodeficiency Virus Type 1-Derived Lentiviral Vectors. Hum Gene Ther Methods 2016; 28:67-77. [PMID: 28042946 DOI: 10.1089/hgtb.2016.150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The use of lentiviral vectors (LVs) for gene transfer in research, technological, or clinical applications requires the production of large amounts of vector. Mass production of clinical-grade LVs remains a challenge and limits certain perspectives for therapeutic use. Some improvements in LV production protocols have been possible by acting on multiple steps of the production process. The addition of animal-derived cholesterol to the culture medium of producer cells is known to increase the infectivity of LVs. To avoid the use of this animal-derived product in clinical settings, an alternative approach is to increase de novo the production of cholesterol by overexpressing a crucial cholesterogenic enzyme, namely, 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR). This project evaluates the impact of such an approach on the production, infectivity, and stability of LVs. We demonstrated that the overexpression of human HMGCR isoform 1 (hHMGCR1) in LV producer cells efficiently increased de novo cholesterol biosynthesis and enhanced by 2- to 3-fold the physical and infectious titers of LVs. We also observed that LVs produced in hHMGCR1-overexpressing cells were comparable in stability to LVs produced under classical conditions and were capable of transducing human CD34+ hematopoietic stem/progenitor cells efficiently. Interestingly, we also showed that LV production in the absence of fetal calf serum (FCS) but under hHMGCR1-overexpressing conditions allowed a viral production yield comparable to that achieved under classical conditions in high FCS content, leading the way to the establishment of new LV production protocols on adherent cells without serum.
Collapse
Affiliation(s)
- Nathalie Holic
- 1 Généthon , Evry, France
- 2 INSERM , UMR_S951, Généthon, Evry, France
- 3 Université Evry Val d'Essonne , UMR_S951, Evry, France
| | - Sophie Frin
- 1 Généthon , Evry, France
- 2 INSERM , UMR_S951, Généthon, Evry, France
| | - Ababacar K Seye
- 1 Généthon , Evry, France
- 2 INSERM , UMR_S951, Généthon, Evry, France
| | - Anne Galy
- 1 Généthon , Evry, France
- 2 INSERM , UMR_S951, Généthon, Evry, France
- 3 Université Evry Val d'Essonne , UMR_S951, Evry, France
| | - David Fenard
- 1 Généthon , Evry, France
- 2 INSERM , UMR_S951, Généthon, Evry, France
- 3 Université Evry Val d'Essonne , UMR_S951, Evry, France
| |
Collapse
|
36
|
Cardile AP, Warren TK, Martins KA, Reisler RB, Bavari S. Will There Be a Cure for Ebola? Annu Rev Pharmacol Toxicol 2016; 57:329-348. [PMID: 27959624 DOI: 10.1146/annurev-pharmtox-010716-105055] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Despite the unprecedented Ebola virus outbreak response in West Africa, no Ebola medical countermeasures have been approved by the US Food and Drug Administration. However, multiple valuable lessons have been learned about the conduct of clinical research in a resource-poor, high risk-pathogen setting. Numerous therapeutics were explored or developed during the outbreak, including repurposed drugs, nucleoside and nucleotide analogues (BCX4430, brincidofovir, favipiravir, and GS-5734), nucleic acid-based drugs (TKM-Ebola and AVI-7537), and immunotherapeutics (convalescent plasma and ZMapp). We review Ebola therapeutics progress in the aftermath of the West Africa Ebola virus outbreak and attempt to offer a glimpse of a path forward.
Collapse
Affiliation(s)
- Anthony P Cardile
- US Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702;
| | - Travis K Warren
- US Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702;
| | - Karen A Martins
- US Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702;
| | - Ronald B Reisler
- US Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702;
| | - Sina Bavari
- US Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702;
| |
Collapse
|
37
|
Balmith M, Faya M, Soliman MES. Ebola virus: A gap in drug design and discovery - experimental and computational perspective. Chem Biol Drug Des 2016; 89:297-308. [PMID: 27637475 DOI: 10.1111/cbdd.12870] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The Ebola virus, formally known as the Ebola hemorrhagic fever, is an acute viral syndrome causing sporadic outbreaks that have ravaged West Africa. Due to its extreme virulence and highly transmissible nature, Ebola has been classified as a category A bioweapon organism. Only recently have vaccine or drug regimens for the Ebola virus been developed, including Zmapp and peptides. In addition, existing drugs which have been repurposed toward anti-Ebola virus activity have been re-examined and are seen to be promising candidates toward combating Ebola. Drug development involving computational tools has been widely employed toward target-based drug design. Screening large libraries have greatly stimulated research toward effective anti-Ebola virus drug regimens. Current emphasis has been placed on the investigation of host proteins and druggable viral targets. There is a huge gap in the literature regarding guidelines in the discovery of Ebola virus inhibitors, which may be due to the lack of information on the Ebola drug targets, binding sites, and mechanism of action of the virus. This review focuses on Ebola virus inhibitors, drugs which could be repurposed to combat the Ebola virus, computational methods which study drug-target interactions as well as providing further insight into the mode of action of the Ebola virus.
Collapse
Affiliation(s)
- Marissa Balmith
- Molecular Modeling and Drug Design Research Group, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Mbuso Faya
- Molecular Modeling and Drug Design Research Group, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Mahmoud E S Soliman
- Molecular Modeling and Drug Design Research Group, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
38
|
Welch SR, Guerrero LW, Chakrabarti AK, McMullan LK, Flint M, Bluemling GR, Painter GR, Nichol ST, Spiropoulou CF, Albariño CG. Lassa and Ebola virus inhibitors identified using minigenome and recombinant virus reporter systems. Antiviral Res 2016; 136:9-18. [PMID: 27771389 DOI: 10.1016/j.antiviral.2016.10.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/11/2016] [Accepted: 10/18/2016] [Indexed: 12/17/2022]
Abstract
Lassa virus (LASV) and Ebola virus (EBOV) infections are important global health issues resulting in significant morbidity and mortality. While several promising drug and vaccine trials for EBOV are ongoing, options for LASV infection are currently limited to ribavirin treatment. A major factor impeding the development of antiviral compounds to treat these infections is the need to manipulate the virus under BSL-4 containment, limiting research to a few institutes worldwide. Here we describe the development of a novel LASV minigenome assay based on the ambisense LASV S segment genome, with authentic terminal untranslated regions flanking a ZsGreen (ZsG) fluorescent reporter protein and a Gaussia princeps luciferase (gLuc) reporter gene. This assay, along with a similar previously established EBOV minigenome, was optimized for high-throughput screening (HTS) of potential antiviral compounds under BSL-2 containment. In addition, we rescued a recombinant LASV expressing ZsG, which, in conjunction with a recombinant EBOV reporter virus, was used to confirm any potential antiviral hits in vitro. Combining an initial screen to identify potential antiviral compounds at BSL-2 containment before progressing to HTS with infectious virus will reduce the amount of expensive and technically challenging BSL-4 containment research. Using these assays, we identified 6-azauridine as having anti-LASV activity, and demonstrated its anti-EBOV activity in human cells. We further identified 2'-deoxy-2'-fluorocytidine as having potent anti-LASV activity, with an EC50 value 10 times lower than that of ribavirin.
Collapse
Affiliation(s)
- Stephen R Welch
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MG G-14, Atlanta, GA, 30329, USA
| | - Lisa Wiggleton Guerrero
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MG G-14, Atlanta, GA, 30329, USA
| | - Ayan K Chakrabarti
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MG G-14, Atlanta, GA, 30329, USA
| | - Laura K McMullan
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MG G-14, Atlanta, GA, 30329, USA
| | - Mike Flint
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MG G-14, Atlanta, GA, 30329, USA
| | | | - George R Painter
- Emory Institute for Drug Development, Emory University, Atlanta, GA, USA
| | - Stuart T Nichol
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MG G-14, Atlanta, GA, 30329, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MG G-14, Atlanta, GA, 30329, USA
| | - César G Albariño
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MG G-14, Atlanta, GA, 30329, USA.
| |
Collapse
|
39
|
Human Survivors of Disease Outbreaks Caused by Ebola or Marburg Virus Exhibit Cross-Reactive and Long-Lived Antibody Responses. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:717-24. [PMID: 27335383 DOI: 10.1128/cvi.00107-16] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/11/2016] [Indexed: 11/20/2022]
Abstract
A detailed understanding of serological immune responses to Ebola and Marburg virus infections will facilitate the development of effective diagnostic methods, therapeutics, and vaccines. We examined antibodies from Ebola or Marburg survivors 1 to 14 years after recovery from disease, by using a microarray that displayed recombinant nucleoprotein (NP), viral protein 40 (VP40), envelope glycoprotein (GP), and inactivated whole virions from six species of filoviruses. All three outbreak cohorts exhibited significant antibody responses to antigens from the original infecting species and a pattern of additional filoviruses that varied by outbreak. NP was the most cross-reactive antigen, while GP was the most specific. Antibodies from survivors of infections by Marburg marburgvirus (MARV) species were least cross-reactive, while those from survivors of infections by Sudan virus (SUDV) species exhibited the highest cross-reactivity. Based on results revealed by the protein microarray, persistent levels of antibodies to GP, NP, and VP40 were maintained for up to 14 years after infection, and survival of infection caused by one species imparted cross-reactive antibody responses to other filoviruses.
Collapse
|
40
|
Jones DM, Padilla-Parra S. The β-Lactamase Assay: Harnessing a FRET Biosensor to Analyse Viral Fusion Mechanisms. SENSORS 2016; 16:s16070950. [PMID: 27347948 PMCID: PMC4970004 DOI: 10.3390/s16070950] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 06/08/2016] [Accepted: 06/20/2016] [Indexed: 02/06/2023]
Abstract
The β-lactamase (BlaM) assay was first revealed in 1998 and was demonstrated to be a robust Förster resonance energy transfer (FRET)-based reporter system that was compatible with a range of commonly-used cell lines. Today, the BlaM assay is available commercially as a kit and can be utilised readily and inexpensively for an array of experimental procedures that require a fluorescence-based readout. One frequent application of the BlaM assay is the measurement of viral fusion—the moment at which the genetic material harboured within virus particles is released into the cytosol following successful entry. The flexibility of the system permits evaluation of not only total fusion levels, but also the kinetics of fusion. However, significant variation exists in the scientific literature regarding the methodology by which the assay is applied to viral fusion analysis, making comparison between results difficult. In this review we draw attention to the disparity of these methodologies and examine the advantages and disadvantages of each approach. Successful strategies shown to render viruses compatible with BlaM-based analyses are also discussed.
Collapse
Affiliation(s)
- Daniel M Jones
- Division of Structural Biology, University of Oxford, The Henry Wellcome Building for Genomic Medicine, Headington, Oxford, OX3 7BN, UK.
| | - Sergi Padilla-Parra
- Division of Structural Biology, University of Oxford, The Henry Wellcome Building for Genomic Medicine, Headington, Oxford, OX3 7BN, UK.
| |
Collapse
|
41
|
Favier AL, Gout E, Reynard O, Ferraris O, Kleman JP, Volchkov V, Peyrefitte C, Thielens NM. Enhancement of Ebola Virus Infection via Ficolin-1 Interaction with the Mucin Domain of GP Glycoprotein. J Virol 2016; 90:5256-5269. [PMID: 26984723 PMCID: PMC4934759 DOI: 10.1128/jvi.00232-16] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/10/2016] [Indexed: 02/05/2023] Open
Abstract
UNLABELLED Ebola virus infection requires the surface viral glycoprotein to initiate entry into the target cells. The trimeric glycoprotein is a highly glycosylated viral protein which has been shown to interact with host C-type lectin receptors and the soluble complement recognition protein mannose-binding lectin, thereby enhancing viral infection. Similarly to mannose-binding lectin, ficolins are soluble effectors of the innate immune system that recognize particular glycans at the pathogen surface. In this study, we demonstrate that ficolin-1 interacts with the Zaire Ebola virus (EBOV) glycoprotein, and we characterized this interaction by surface plasmon resonance spectroscopy. Ficolin-1 was shown to bind to the viral glycoprotein with a high affinity. This interaction was mediated by the fibrinogen-like recognition domain of ficolin-1 and the mucin-like domain of the viral glycoprotein. Using a ficolin-1 control mutant devoid of sialic acid-binding capacity, we identified sialylated moieties of the mucin domain to be potential ligands on the glycoprotein. In cell culture, using both pseudotyped viruses and EBOV, ficolin-1 was shown to enhance EBOV infection independently of the serum complement. We also observed that ficolin-1 enhanced EBOV infection on human monocyte-derived macrophages, described to be major viral target cells,. Competition experiments suggested that although ficolin-1 and mannose-binding lectin recognized different carbohydrate moieties on the EBOV glycoprotein, the observed enhancement of the infection likely depended on a common cellular receptor/partner. In conclusion, ficolin-1 could provide an alternative receptor-mediated mechanism for enhancing EBOV infection, thereby contributing to viral subversion of the host innate immune system. IMPORTANCE A specific interaction involving ficolin-1 (M-ficolin), a soluble effector of the innate immune response, and the glycoprotein (GP) of EBOV was identified. Ficolin-1 enhanced virus infection instead of tipping the balance toward its elimination. An interaction between the fibrinogen-like recognition domain of ficolin-1 and the mucin-like domain of Ebola virus GP occurred. In this model, the enhancement of infection was shown to be independent of the serum complement. The facilitation of EBOV entry into target host cells by the interaction with ficolin-1 and other host lectins shunts virus elimination, which likely facilitates the survival of the virus in infected host cells and contributes to the virus strategy to subvert the innate immune response.
Collapse
Affiliation(s)
- Anne-Laure Favier
- Unité de Virologie, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Evelyne Gout
- Université Grenoble Alpes, Grenoble, France
- CNRS, IBS, Grenoble, France
- CEA, IBS, Grenoble, France
| | - Olivier Reynard
- Molecular Basis of Viral Pathogenicity, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111-CNRS UMR5308, Université Lyon 1, ENS de Lyon, Lyon, France
| | - Olivier Ferraris
- Unité de Virologie, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Jean-Philippe Kleman
- Université Grenoble Alpes, Grenoble, France
- CNRS, IBS, Grenoble, France
- CEA, IBS, Grenoble, France
| | - Viktor Volchkov
- Molecular Basis of Viral Pathogenicity, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111-CNRS UMR5308, Université Lyon 1, ENS de Lyon, Lyon, France
| | - Christophe Peyrefitte
- Unité de Virologie, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Nicole M Thielens
- Université Grenoble Alpes, Grenoble, France
- CNRS, IBS, Grenoble, France
- CEA, IBS, Grenoble, France
| |
Collapse
|
42
|
Beck S, Henß L, Weidner T, Herrmann J, Müller R, Chao YK, Grimm C, Weber C, Sliva K, Schnierle BS. Identification of entry inhibitors of Ebola virus pseudotyped vectors from a myxobacterial compound library. Antiviral Res 2016; 132:85-91. [PMID: 27241689 DOI: 10.1016/j.antiviral.2016.05.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/11/2016] [Accepted: 05/24/2016] [Indexed: 11/29/2022]
Abstract
Myxobacteria produce secondary metabolites many of which were described to have various biological effects including anti-fungal, anti-bacterial and anti-viral activity. The majority of these metabolites are novel scaffolds with unique modes-of-action and hence might be potential leads for drug discovery. Here, we tested a myxobacterial natural product library for compounds with inhibitory activity against Ebola virus (EBOV). The assay was performed with a surrogate system using Ebola envelope glycoprotein (GP) pseudotyped lentiviral vectors. EBOV specificity was proven by counter-screening with vesicular stomatitis virus G protein pseudotyped vectors. Two compounds were identified that preferentially inhibited EBOV GP mediated cell entry: Chondramides that act on the actin skeleton but might be too toxic and noricumazole A, a potassium channel inhibitor, which might constitute a novel pathway to inhibit Ebola virus cell entry.
Collapse
Affiliation(s)
- Simon Beck
- Paul-Ehrlich-Institut, Department of Virology, Paul-Ehrlich Strasse 51-59, 63225 Langen, Germany
| | - Lisa Henß
- Paul-Ehrlich-Institut, Department of Virology, Paul-Ehrlich Strasse 51-59, 63225 Langen, Germany
| | - Tatjana Weidner
- Paul-Ehrlich-Institut, Department of Virology, Paul-Ehrlich Strasse 51-59, 63225 Langen, Germany
| | - Jennifer Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland, Microbial Natural Products, Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland, Microbial Natural Products, Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany
| | - Yu-Kai Chao
- Department of Pharmacy - Center for Drug Research and Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilians-Universität München, Germany
| | - Christian Grimm
- Department of Pharmacy - Center for Drug Research and Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilians-Universität München, Germany
| | - Christopher Weber
- Paul-Ehrlich-Institut, Department of Virology, Paul-Ehrlich Strasse 51-59, 63225 Langen, Germany
| | - Katja Sliva
- Paul-Ehrlich-Institut, Department of Virology, Paul-Ehrlich Strasse 51-59, 63225 Langen, Germany
| | - Barbara S Schnierle
- Paul-Ehrlich-Institut, Department of Virology, Paul-Ehrlich Strasse 51-59, 63225 Langen, Germany.
| |
Collapse
|
43
|
Lee J, Gregory SM, Nelson EA, White JM, Tamm LK. The Roles of Histidines and Charged Residues as Potential Triggers of a Conformational Change in the Fusion Loop of Ebola Virus Glycoprotein. PLoS One 2016; 11:e0152527. [PMID: 27023721 PMCID: PMC4811418 DOI: 10.1371/journal.pone.0152527] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/15/2016] [Indexed: 11/23/2022] Open
Abstract
Ebola virus (EBOV) enters cells from late endosomes/lysosomes under mildly acidic conditions. Entry by fusion with the endosomal membrane requires the fusion loop (FL, residues 507–560) of the EBOV surface glycoprotein to undergo a pH-dependent conformational change. To find the pH trigger for this reaction we mutated multiple conserved histidines and charged and uncharged hydrophilic residues in the FL and measured their activity by liposome fusion and cell entry of virus-like particles. The FL location in the membrane was assessed by NMR using soluble and lipid-bound paramagnetic relaxation agents. While we could not identify a single residue to be alone responsible for pH triggering, we propose that a distributed pH effect over multiple residues induces the conformational change that enhances membrane insertion and triggers the fusion activity of the EBOV FL.
Collapse
Affiliation(s)
- Jinwoo Lee
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia 22908, United States of America.,Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22908, United States of America
| | - Sonia M Gregory
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia 22908, United States of America.,Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22908, United States of America
| | - Elizabeth A Nelson
- Department Cell Biology, University of Virginia, Charlottesville, Virginia 22908, United States of America
| | - Judith M White
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia 22908, United States of America.,Department Cell Biology, University of Virginia, Charlottesville, Virginia 22908, United States of America
| | - Lukas K Tamm
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, Virginia 22908, United States of America.,Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22908, United States of America
| |
Collapse
|
44
|
Antanasijevic A, Kingsley C, Basu A, Bowlin TL, Rong L, Caffrey M. Application of virus-like particles (VLP) to NMR characterization of viral membrane protein interactions. JOURNAL OF BIOMOLECULAR NMR 2016; 64:255-65. [PMID: 26921030 PMCID: PMC4826305 DOI: 10.1007/s10858-016-0025-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/22/2016] [Indexed: 05/10/2023]
Abstract
The membrane proteins of viruses play critical roles in the virus life cycle and are attractive targets for therapeutic intervention. Virus-like particles (VLP) present the possibility to study the biochemical and biophysical properties of viral membrane proteins in their native environment. Specifically, the VLP constructs contain the entire protein sequence and are comprised of native membrane components including lipids, cholesterol, carbohydrates and cellular proteins. In this study we prepare VLP containing full-length hemagglutinin (HA) or neuraminidase (NA) from influenza and characterize their interactions with small molecule inhibitors. Using HA-VLP, we first show that VLP samples prepared using the standard sucrose gradient purification scheme contain significant amounts of serum proteins, which exhibit high potential for non-specific interactions, thereby complicating NMR studies of ligand-target interactions. We then show that the serum contaminants may be largely removed with the addition of a gel filtration chromatography step. Next, using HA-VLP we demonstrate that WaterLOGSY NMR is significantly more sensitive than Saturation Transfer Difference (STD) NMR for the study of ligand interactions with membrane bound targets. In addition, we compare the ligand orientation to HA embedded in VLP with that of recombinant HA by STD NMR. In a subsequent step, using NA-VLP we characterize the kinetic and binding properties of substrate analogs and inhibitors of NA, including study of the H274Y-NA mutant, which leads to wide spread resistance to current influenza antivirals. In summary, our work suggests that VLP have high potential to become standard tools in biochemical and biophysical studies of viral membrane proteins, particularly when VLP are highly purified and combined with control VLP containing native membrane proteins.
Collapse
Affiliation(s)
- Aleksandar Antanasijevic
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland, Chicago, IL, 60607, USA
| | - Carolyn Kingsley
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland, Chicago, IL, 60607, USA
| | - Arnab Basu
- Microbiotix Inc., Worcester, MA, 01605, USA
| | | | - Lijun Rong
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Michael Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland, Chicago, IL, 60607, USA.
| |
Collapse
|
45
|
Zhang X, Ao Z, Bello A, Ran X, Liu S, Wigle J, Kobinger G, Yao X. Characterization of the inhibitory effect of an extract of Prunella vulgaris on Ebola virus glycoprotein (GP)-mediated virus entry and infection. Antiviral Res 2016; 127:20-31. [PMID: 26778707 PMCID: PMC7113790 DOI: 10.1016/j.antiviral.2016.01.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 01/05/2016] [Accepted: 01/06/2016] [Indexed: 11/25/2022]
Abstract
Currently, no approved antiviral therapeutic is available for treatment or prevention of Ebola virus (EBOV) infection. In this study, we characterized an EBOV-glycoprotein (GP) pseudotyped HIV-1-based vector system in different cell cultures, including human umbilical vein endothelial cells (HUVECs) and human macrophages, for the screening of anti-EBOV-GP agent(s). Based on this system, we demonstrated that an aqueous extract (CHPV) from the Chinese herb Prunella vulgaris displayed a potent inhibitory effect on EBOV-GP pseudotyped virus (EBOV-GP-V)-mediated infection in various cell lines, including HUVEC and macrophage. In addition, our results indicated that CHPV was able to block an eGFP-expressing Zaire ebola virus (eGFP-ZEBOV) infection in VeroE6 cells. The anti-EBOV activity of CHPV was exhibited in a dose-dependent manner. At a 12.5 μg/ml concentration, the CHPV showed a greater than 80% inhibition of EBOV-GP-V and eGFP-EBOV infections. Likewise, our studies suggested that the inhibitory effect of CHPV occurred by binding directly to EBOV-GP-Vs and blocking the early viral events. Interestingly, our results have shown that CHPV was able to enhance the anti-EBOV activity of the monoclonal antibody MAb 2G4 against EBOV-GP. Overall, this study provides evidence that CHPV has anti-EBOV activity and may be developed as a novel antiviral approach against EBOV infection.
Collapse
Affiliation(s)
- Xu Zhang
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Canada
| | - Zhujun Ao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Canada; Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, 410078, PR China
| | - Alexander Bello
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Canada
| | - Xiaozhuo Ran
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Canada
| | - Shuiping Liu
- Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, 410078, PR China
| | - Jeffrey Wigle
- Department of Biochemistry and Medical Genetics, University of Manitoba, Canada
| | - Gary Kobinger
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Canada
| | - Xiaojian Yao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Canada; Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, 410078, PR China.
| |
Collapse
|
46
|
Miao C, Li M, Zheng YM, Cohen FS, Liu SL. Cell-cell contact promotes Ebola virus GP-mediated infection. Virology 2015; 488:202-15. [PMID: 26655238 DOI: 10.1016/j.virol.2015.11.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 11/18/2015] [Accepted: 11/19/2015] [Indexed: 10/22/2022]
Abstract
Ebola virus (EBOV) is a highly pathogenic filovirus that causes hemorrhagic fever in humans and animals. Here we provide evidence that cell-cell contact promotes infection mediated by the glycoprotein (GP) of EBOV. Interestingly, expression of EBOV GP alone, even in the absence of retroviral Gag-Pol, is sufficient to transfer a retroviral vector encoding Tet-off from cell to cell. Cell-to-cell infection mediated by EBOV GP is blocked by inhibitors of actin polymerization, but appears to be less sensitive to KZ52 neutralization. Treatment of co-cultured cells with cathepsin B/L inhibitors, or an entry inhibitor 3.47 that targets the receptor NPC1 for virus binding, also blocks cell-to-cell infection. Cell-cell contact also enhances spread of rVSV bearing GP in monocytes and macrophages, the primary targets of natural EBOV infection. Altogether, our study reveals that cell-cell contact promotes EBOV GP-mediated infection, and provides new insight into understanding EBOV spread and viral pathogenesis.
Collapse
Affiliation(s)
- Chunhui Miao
- Department of Molecular Microbiology and Immunology, Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Minghua Li
- Department of Molecular Microbiology and Immunology, Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Yi-Min Zheng
- Department of Molecular Microbiology and Immunology, Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Fredric S Cohen
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Shan-Lu Liu
- Department of Molecular Microbiology and Immunology, Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA.
| |
Collapse
|
47
|
Messaoudi I, Amarasinghe GK, Basler CF. Filovirus pathogenesis and immune evasion: insights from Ebola virus and Marburg virus. Nat Rev Microbiol 2015; 13:663-76. [PMID: 26439085 DOI: 10.1038/nrmicro3524] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Ebola viruses and Marburg viruses, members of the filovirus family, are zoonotic pathogens that cause severe disease in people, as highlighted by the latest Ebola virus epidemic in West Africa. Filovirus disease is characterized by uncontrolled virus replication and the activation of host responses that contribute to pathogenesis. Underlying these phenomena is the potent suppression of host innate antiviral responses, particularly the type I interferon response, by viral proteins, which allows high levels of viral replication. In this Review, we describe the mechanisms used by filoviruses to block host innate immunity and discuss the links between immune evasion and filovirus pathogenesis.
Collapse
Affiliation(s)
- Ilhem Messaoudi
- School of Medicine, University of California Riverside, Riverside, California 92521, USA
| | - Gaya K Amarasinghe
- The Division of Biology &Biomedical Sciences, Washington University in St. Louis, St. Louis, Missouri 63110, USA
| | - Christopher F Basler
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
48
|
Abstract
Ebola virus disease (EVD) is a zoonotic disease that causes severe haemorrhagic fever, with high fatality rates of up to 90% in humans. Today, there is no effective treatment available. Person-to-person transmission occurs through exposure to blood or body fluids, which can threaten other household members and first-line healthcare workers. The first cases of EVD in Guinea were identified on 22 March 2014. It was initially believed that this like previous outbreaks would be self-limiting. However, lack of public health infrastructure, delays in virus detection and late implementation of control interventions contributed to widespread transmission of EVD in a region inexperienced in dealing with the disease. Socio-cultural and economic factors probably also played a key role in the spread of the disease, resulting in the current large-scale outbreak. Some promising candidate treatments for this disease are now being developed.
Collapse
Affiliation(s)
- A Mirazimi
- Department of Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden
- National Veterinary Institute, Uppsala, Sweden
| |
Collapse
|
49
|
Jones DM, Padilla-Parra S. Imaging real-time HIV-1 virion fusion with FRET-based biosensors. Sci Rep 2015; 5:13449. [PMID: 26300212 PMCID: PMC4547133 DOI: 10.1038/srep13449] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 07/28/2015] [Indexed: 11/08/2022] Open
Abstract
We have produced a novel, simple and rapid method utilising genetically encodable FRET-based biosensors to permit the detection of HIV-1 virion fusion in living cells. These biosensors show high sensitivity both spatially and temporally, and allow the real-time recovery of HIV-1 fusion kinetics in both single cells and cell populations simultaneously.
Collapse
Affiliation(s)
- Daniel M. Jones
- Division of Structural Biology, University of Oxford, The Henry Wellcome Building for Genomic Medicine, Headington, Oxford OX3 7BN, UK
| | - Sergi Padilla-Parra
- Division of Structural Biology, University of Oxford, The Henry Wellcome Building for Genomic Medicine, Headington, Oxford OX3 7BN, UK
- Cellular Imaging Core, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
50
|
Janeba Z. Development of Small-Molecule Antivirals for Ebola. Med Res Rev 2015; 35:1175-94. [PMID: 26172225 PMCID: PMC7168439 DOI: 10.1002/med.21355] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 05/21/2015] [Accepted: 05/23/2015] [Indexed: 01/05/2023]
Abstract
Ebola hemorrhagic fever is a deadly disease caused by infection with one of the Ebola virus species. Although a significant progress has recently been made in understanding of Ebola virus biology and pathogenesis, development of effective anti-Ebola treatments has not been very productive, compared to other areas of antiviral research (e.g., HIV and HCV infections). No approved vaccine or medicine is available for Ebola but several are currently under development. This review summarises attempts in identification, evaluation, and development of small-molecule candidates for treatment of Ebola viral disease, including the most promising experimental drugs brincidofovir (CMX001), BCX4430, and favipiravir (T-705).
Collapse
Affiliation(s)
- Zlatko Janeba
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i. Flemingovo nám. 2, CZ-16610 Prague 6, Czech Republic
| |
Collapse
|