1
|
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
|
2
|
Marx de Andrade R, Paulos A, Valadas E, Laerte Pinto Junior V. [Disease Caused by Filoviruses: An Update]. ACTA MEDICA PORT 2025; 38:42-48. [PMID: 39566016 DOI: 10.20344/amp.21911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/24/2024] [Indexed: 11/22/2024]
Abstract
The Marburg and Ebola viruses belong to the Filoviridae family and are known to cause emerging zoonotic diseases. These viruses have a high case fatality rate and are easily transmissible from person to person, which makes them capable of triggering outbreaks, including in non-endemic regions, and are also considered agents of bioterrorism. Fruit bats are the natural reservoirs of these filoviruses. Transmission to humans occurs through direct contact with bodily fluids or tissues from infected animals or humans. The most severe form of filovirus disease manifests as mucocutaneous hemorrhage, often accompanied by multiorgan failure, which is the main cause of death. Traditionally, these diseases are classified in the group of viral hemorrhagic fevers, although this term is being abandoned, as there are not always hemorrhagic manifestations or fever in the patient's clinical history. Currently, no specific antiviral treatment for filovirus disease exists, and the therapeutic approach consists of supportive measures. However, for the Zaire Ebola virus (EBOV), monoclonal antibodies have already been licensed for treatment and post-exposure prophylaxis, in addition to three vaccines available. Due to the public health importance and the possibility of cases outside Africa, this review aims to improve clinical knowledge and the approach to suspected cases of FD. Improved surveillance and preparedness for potential global outbreaks are essential measures to effectively respond to these public health threats and to ensure that healthcare professionals are well-informed and prepared to deal with these diseases.
Collapse
Affiliation(s)
| | | | - Emília Valadas
- Clínica Universitária de Doenças Infecciosas. Faculdade de Medicina. Universidade de Lisboa. Lisboa. Portugal
| | - Vitor Laerte Pinto Junior
- Faculdade de Medicina. Universidade de Lisboa. Lisboa; Unidade Local de Saúde da Arrábida. Setúbal. Portugal
| |
Collapse
|
3
|
Sanford LM, Keiser P, Fujii N, Woods H, Zhang C, Xu Z, Mahajani NS, Cortés JG, Plescia CB, Knipp G, Stahelin RV, Davey R, Davisson VJ. Evaluation of potency and metabolic stability of diphyllin-derived Vacuolar-ATPase inhibitors. Eur J Med Chem 2024; 275:116537. [PMID: 38875806 PMCID: PMC11236507 DOI: 10.1016/j.ejmech.2024.116537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/16/2024] [Accepted: 05/26/2024] [Indexed: 06/16/2024]
Abstract
Diphyllin is a naturally occurring lignan comprised of an aryl naphthalene lactone scaffold that demonstrates beneficial biological activities in disease models of cancer, obesity, and viral infection. A target of diphyllin and naturally occurring derivatives is the vacuolar ATPase (V-ATPase) complex. Although diphyllin-related natural products are active with in vitro models for viral entry, the potencies and unknown pharmacokinetic properties limit well-designed in vivo evaluations. Previous studies demonstrated that diphyllin derivatives have the utility of blocking the Ebola virus cell entry pathway. However, diphyllin shows limited potency and poor oral bioavailability in mice. An avenue to improve the potency was used in a new library of synthetic derivatives of diphyllin. Diphyllin derivatives exploiting ether linkages at the 4-position with one-to-three carbon spacers to an oxygen or nitrogen atom provided compounds with EC50 values ranging from 7 to 600 nM potency and selectivity up to >500 against Ebola virus in infection assays. These relative potencies are reflected in the Ebola virus infection of primary macrophages, a cell type involved in early pathogenesis. A target engagement study reveals that reducing the ATPV0a2 protein expression enhanced the potency of diphyllin derivatives to block EBOV entry, consistent with effects on the endosomal V-ATPase function. Despite the substantial enhancement of antiviral potencies, limitations were identified, including rapid clearance predicted by in vitro microsome stability assays. However, compounds with similar or improved half-lives relative to diphyllin demonstrated improved pharmacokinetic profiles in vivo. Importantly, these derivatives displayed suitable plasma levels using oral administration, establishing the feasibility of in vivo antiviral testing.
Collapse
Affiliation(s)
- Laura M Sanford
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, United States
| | - Patrick Keiser
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02118, United States
| | - Naoaki Fujii
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, United States
| | - Hannah Woods
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, United States
| | - Charlie Zhang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, United States; Department of Industrial and Molecular Pharmaceutics, Purdue University, West Lafayette, IN, 47907, United States
| | - Zhuangyan Xu
- Department of Industrial and Molecular Pharmaceutics, Purdue University, West Lafayette, IN, 47907, United States
| | - Nivedita S Mahajani
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, United States
| | - Julián González Cortés
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, United States
| | - Caroline B Plescia
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, United States
| | - Gregory Knipp
- Department of Industrial and Molecular Pharmaceutics, Purdue University, West Lafayette, IN, 47907, United States
| | - Robert V Stahelin
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, United States
| | - Robert Davey
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02118, United States
| | - Vincent Jo Davisson
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, United States.
| |
Collapse
|
4
|
Durante D, Bott R, Cooper L, Owen C, Morsheimer KM, Patten JJ, Zielinski C, Peet NP, Davey RA, Gaisina IN, Rong L, Moore TW. N-Substituted Pyrrole-Based Heterocycles as Broad-Spectrum Filoviral Entry Inhibitors. J Med Chem 2024; 67:13737-13764. [PMID: 39169825 DOI: 10.1021/acs.jmedchem.4c00527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Since the largest and most fatal Ebola virus epidemic during 2014-2016, there have been several consecutive filoviral outbreaks in recent years, including those in 2021, 2022, and 2023. Ongoing outbreak prevalence and limited FDA-approved filoviral therapeutics emphasize the need for novel small molecule treatments. Here, we showcase the structure-activity relationship development of N-substituted pyrrole-based heterocycles and their potent, submicromolar entry inhibition against diverse filoviruses in a target-based pseudovirus assay. Inhibitor antiviral activity was validated using replication-competent Ebola, Sudan, and Marburg viruses. Mutational analysis was used to map the targeted region within the Ebola virus glycoprotein. Antiviral counter-screen and phospholipidosis assays were performed to demonstrate the reduced off-target activity of these filoviral entry inhibitors. Favorable antiviral potency, selectivity, and drug-like properties of the N-substituted pyrrole-based heterocycles support their potential as broad-spectrum antifiloviral treatments.
Collapse
Affiliation(s)
- Destiny Durante
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Ryan Bott
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Laura Cooper
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Callum Owen
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Medical Campus, Boston, Massachusetts 02118, United States
| | - Kimberly M Morsheimer
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Medical Campus, Boston, Massachusetts 02118, United States
| | - J J Patten
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Medical Campus, Boston, Massachusetts 02118, United States
| | - Christian Zielinski
- UICentre: Drug Discovery, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Norton P Peet
- Chicago BioSolutions Inc., Chicago, Illinois 60612, United States
| | - Robert A Davey
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Medical Campus, Boston, Massachusetts 02118, United States
| | - Irina N Gaisina
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, Illinois 60612, United States
- Chicago BioSolutions Inc., Chicago, Illinois 60612, United States
- UICentre: Drug Discovery, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Lijun Rong
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois 60612, United States
- Chicago BioSolutions Inc., Chicago, Illinois 60612, United States
| | - Terry W Moore
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, Illinois 60612, United States
- University of Illinois Cancer Center, University of Illinois Chicago, Chicago, Illinois 60612, United States
| |
Collapse
|
5
|
Strebinger D, Frangieh CJ, Friedrich MJ, Faure G, Macrae RK, Zhang F. Cell type-specific delivery by modular envelope design. Nat Commun 2023; 14:5141. [PMID: 37612276 PMCID: PMC10447438 DOI: 10.1038/s41467-023-40788-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 08/10/2023] [Indexed: 08/25/2023] Open
Abstract
The delivery of genetic cargo remains one of the largest obstacles to the successful translation of experimental therapies, in large part due to the absence of targetable delivery vectors. Enveloped delivery modalities use viral envelope proteins, which determine tropism and induce membrane fusion. Here we develop DIRECTED (Delivery to Intended REcipient Cells Through Envelope Design), a modular platform that consists of separate fusion and targeting components. To achieve high modularity and programmable cell type specificity, we develop multiple strategies to recruit or immobilize antibodies on the viral envelope, including a chimeric antibody binding protein and a SNAP-tag enabling the use of antibodies or other proteins as targeting molecules. Moreover, we show that fusogens from multiple viral families are compatible with DIRECTED and that DIRECTED components can target multiple delivery chassis (e.g., lentivirus and MMLV gag) to specific cell types, including primary human T cells in PBMCs and whole blood.
Collapse
Affiliation(s)
- Daniel Strebinger
- Howard Hughes Medical Institute, Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Chris J Frangieh
- Howard Hughes Medical Institute, Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Mirco J Friedrich
- Howard Hughes Medical Institute, Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Guilhem Faure
- Howard Hughes Medical Institute, Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Rhiannon K Macrae
- Howard Hughes Medical Institute, Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Feng Zhang
- Howard Hughes Medical Institute, Cambridge, MA, 02139, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
6
|
Abir MH, Rahman T, Das A, Etu SN, Nafiz IH, Rakib A, Mitra S, Emran TB, Dhama K, Islam A, Siyadatpanah A, Mahmud S, Kim B, Hassan MM. Pathogenicity and virulence of Marburg virus. Virulence 2022; 13:609-633. [PMID: 35363588 PMCID: PMC8986239 DOI: 10.1080/21505594.2022.2054760] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/10/2022] [Accepted: 03/13/2022] [Indexed: 12/25/2022] Open
Abstract
Marburg virus (MARV) has been a major concern since 1967, with two major outbreaks occurring in 1998 and 2004. Infection from MARV results in severe hemorrhagic fever, causing organ dysfunction and death. Exposure to fruit bats in caves and mines, and human-to-human transmission had major roles in the amplification of MARV outbreaks in African countries. The high fatality rate of up to 90% demands the broad study of MARV diseases (MVD) that correspond with MARV infection. Since large outbreaks are rare for MARV, clinical investigations are often inadequate for providing the substantial data necessary to determine the treatment of MARV disease. Therefore, an overall review may contribute to minimizing the limitations associated with future medical research and improve the clinical management of MVD. In this review, we sought to analyze and amalgamate significant information regarding MARV disease epidemics, pathophysiology, and management approaches to provide a better understanding of this deadly virus and the associated infection.
Collapse
Affiliation(s)
- Mehedy Hasan Abir
- Faculty of Food Science and Technology, Chattogram Veterinary and Animal Sciences University, Chittagong, Bangladesh
| | - Tanjilur Rahman
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Ayan Das
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Silvia Naznin Etu
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Iqbal Hossain Nafiz
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Ahmed Rakib
- Department of Pharmacy, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Ariful Islam
- EcoHealth Alliance, New York, NY, USA
- Centre for Integrative Ecology, School of Life and Environmental Science, Deakin University, Victoria, Australia
| | - Abolghasem Siyadatpanah
- Ferdows School of Paramedical and Health, Birjand University of Medical Sciences, Birjand, Iran
| | - Shafi Mahmud
- Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | - Bonlgee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Mohammad Mahmudul Hassan
- Queensland Alliance for One Health Sciences, School of Veterinary Sciences, The University of Queensland, Gatton, Australia
- Department of Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University, Chattogram, Bangladesh
| |
Collapse
|
7
|
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
|
8
|
Abstract
Varicella-zoster virus (VZV) maintains lifelong latency in neurons following initial infection and can subsequently be reactivated to result in herpes zoster or severe neurological manifestations such as encephalitis. Mechanisms of VZV neuropathogenesis have been challenging to study due to the strict human tropism of the virus. Although neuronal entry mediators of other herpesviruses, including herpes simplex virus, have been identified, little is known regarding how VZV enters neurons. Here, we utilize a human stem cell-based neuronal model to characterize cellular factors that mediate entry. Through transcriptional profiling of infected cells, we identify the cell adhesion molecule nectin-1 as a candidate mediator of VZV entry. Nectin-1 is highly expressed in the cell bodies and axons of neurons. Either knockdown of endogenous nectin-1 or incubation with soluble forms of nectin-1 produced in mammalian cells results in a marked decrease in infectivity of neurons. Notably, while addition of soluble nectin-1 during viral infection inhibits infectivity, addition after infection has no effect on infectivity. Ectopic expression of human nectin-1 in a cell line resistant to productive VZV infection confers susceptibility to infection. In summary, we have identified nectin-1 as a neuronal entry mediator of VZV. IMPORTANCE Varicella-zoster virus (VZV) causes chickenpox, gains access to neurons during primary infection where it resides lifelong, and can later be reactivated. Reactivation is associated with shingles and postherpetic neuralgia, as well as with severe neurologic complications, including vasculitis and encephalitis. Although the varicella vaccine substantially decreases morbidity and mortality associated with primary infection, the vaccine cannot prevent the development of neuronal latency, and vaccinated populations are still at risk for reactivation. Furthermore, immunocompromised individuals are at higher risk for VZV reactivation and associated complications. Little is known regarding how VZV enters neurons. Here, we identify nectin-1 as an entry mediator of VZV in human neurons. Identification of nectin-1 as a neuronal VZV entry mediator could lead to improved treatments and preventative measures to reduce VZV related morbidity and mortality.
Collapse
|
9
|
Modeling the efficiency of filovirus entry into cells in vitro: Effects of SNP mutations in the receptor molecule. PLoS Comput Biol 2020; 16:e1007612. [PMID: 32986692 PMCID: PMC7544041 DOI: 10.1371/journal.pcbi.1007612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 10/08/2020] [Accepted: 08/03/2020] [Indexed: 11/27/2022] Open
Abstract
Interaction between filovirus glycoprotein (GP) and the Niemann-Pick C1 (NPC1) protein is essential for membrane fusion during virus entry. Some single-nucleotide polymorphism (SNPs) in two surface-exposed loops of NPC1 are known to reduce viral infectivity. However, the dependence of differences in entry efficiency on SNPs remains unclear. Using vesicular stomatitis virus pseudotyped with Ebola and Marburg virus GPs, we investigated the cell-to-cell spread of viruses in cultured cells expressing NPC1 or SNP derivatives. Eclipse and virus-producing phases were assessed by in vitro infection experiments, and we developed a mathematical model describing spatial-temporal virus spread. This mathematical model fit the plaque radius data well from day 2 to day 6. Based on the estimated parameters, we found that SNPs causing the P424A and D508N substitutions in NPC1 most effectively reduced the entry efficiency of Ebola and Marburg viruses, respectively. Our novel approach could be broadly applied to other virus plaque assays. Ebola (EBOV) and Marburg (MARV) viruses, which are included viruses of the family Filoviridae, cause severe hemorrhagic fever in humans. Filovirus particles is adsorbed to the cell through glycoprotein (GP), which is the only viral surface protein. Interaction between the filovirus sugar protein (GP) and the Niemann-Pick C1 (NPC1) protein plays a key role in membrane fusion during virus entry. Although some single-nucleotide polymorphism (SNPs) in two surface-exposed loops of NPC1 are known to reduce viral infectivity, the dependence of differences in entry efficiency on SNPs has not been studied. We therefore investigated the cell-to-cell spread of viruses in cultured cells expressing NPC1 or SNP derivatives. Using a mathematical model describing spatial-temporal virus spread, we quantitatively analyze viral entry efficiency and how this affected cell-to-cell spread. Our approach may be applied to not only understanding the roles of genetic polymorphisms in human susceptibility to filoviruses, but also other virus plaque assays.
Collapse
|
10
|
Davey RA, Shtanko O, Anantpadma M, Sakurai Y, Chandran K, Maury W. Mechanisms of Filovirus Entry. Curr Top Microbiol Immunol 2017; 411:323-352. [PMID: 28601947 DOI: 10.1007/82_2017_14] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Filovirus entry into cells is complex, perhaps as complex as any viral entry mechanism identified to date. However, over the past 10 years, the important events required for filoviruses to enter into the endosomal compartment and fuse with vesicular membranes have been elucidated (Fig. 1). Here, we highlight the important steps that are required for productive entry of filoviruses into mammalian cells.
Collapse
Affiliation(s)
- R A Davey
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - O Shtanko
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - M Anantpadma
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Y Sakurai
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - K Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - W Maury
- Department of Microbiology, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
11
|
Lawrence P, Danet N, Reynard O, Volchkova V, Volchkov V. Human transmission of Ebola virus. Curr Opin Virol 2016; 22:51-58. [PMID: 28012412 DOI: 10.1016/j.coviro.2016.11.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/25/2016] [Accepted: 11/29/2016] [Indexed: 12/11/2022]
Abstract
Ever since the first recognised outbreak of Ebolavirus in 1976, retrospective epidemiological analyses and extensive studies with animal models have given us insight into the nature of the pathology and transmission mechanisms of this virus. In this review focusing on Ebolavirus, we present an outline of our current understanding of filovirus human-to-human transmission and of our knowledge concerning the molecular basis of viral transmission and potential for adaptation, with particular focus on what we have learnt from the 2014 outbreak in West Africa. We identify knowledge gaps relating to transmission and pathogenicity mechanisms, molecular adaptation and filovirus ecology.
Collapse
Affiliation(s)
- Philip Lawrence
- Molecular Basis of Viral Pathogenicity, International Centre for Research in Infectiology (CIRI), INSERM U1111 - CNRS UMR5308, Université Lyon 1, Ecole Normale Supérieure de Lyon, Lyon 69007, France; Université de Lyon, UMRS 449, Laboratoire de Biologie Générale, Université Catholique de Lyon - EPHE, Lyon 69288, France
| | - Nicolas Danet
- Molecular Basis of Viral Pathogenicity, International Centre for Research in Infectiology (CIRI), INSERM U1111 - CNRS UMR5308, Université Lyon 1, Ecole Normale Supérieure de Lyon, Lyon 69007, France
| | - Olivier Reynard
- Molecular Basis of Viral Pathogenicity, International Centre for Research in Infectiology (CIRI), INSERM U1111 - CNRS UMR5308, Université Lyon 1, Ecole Normale Supérieure de Lyon, Lyon 69007, France
| | - Valentina Volchkova
- Molecular Basis of Viral Pathogenicity, International Centre for Research in Infectiology (CIRI), INSERM U1111 - CNRS UMR5308, Université Lyon 1, Ecole Normale Supérieure de Lyon, Lyon 69007, France
| | - Viktor Volchkov
- Molecular Basis of Viral Pathogenicity, International Centre for Research in Infectiology (CIRI), INSERM U1111 - CNRS UMR5308, Université Lyon 1, Ecole Normale Supérieure de Lyon, Lyon 69007, France.
| |
Collapse
|
12
|
Filovirus proteins for antiviral drug discovery: A structure/function analysis of surface glycoproteins and virus entry. Antiviral Res 2016; 135:1-14. [PMID: 27640102 PMCID: PMC7113884 DOI: 10.1016/j.antiviral.2016.09.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 09/01/2016] [Accepted: 09/05/2016] [Indexed: 12/20/2022]
Abstract
This review focuses on the recent progress in our understanding of filovirus protein structure/function and its impact on antiviral research. Here we focus on the surface glycoprotein GP1,2 and its different roles in filovirus entry. We first describe the latest advances on the characterization of GP gene-overlapping proteins sGP, ssGP and Δ-peptide. Then, we compare filovirus surface GP1,2 proteins in terms of structure, synthesis and function. As they bear potential in drug-design, the discovery of small organic compounds inhibiting filovirus entry is a currently very active field. Although it is at an early stage, the development of antiviral drugs against Ebola and Marburg virus entry might prove essential to reduce outbreak-associated fatality rates through post-exposure treatment of both suspected and confirmed cases. The filovirus surface glycoprotein is the key player protein responsible for viral entry. Secreted forms of the glycoprotein have been suggested to participate to filovirus virus pathogenicity. Recent structural insights of the filovirus surface glycoprotein highlight new antiviral perspectives. Interesting compounds and innovative antiviral strategies emerge from research and development to inhibit filovirus entry.
Collapse
|
13
|
Inhibitory Antibodies Targeting Emerging Viruses: Advancements and Mechanisms. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:535-9. [PMID: 27226280 DOI: 10.1128/cvi.00136-16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
From Ebola virus outbreaks in Western Africa to the introduction of chikungunya and Zika viruses in the Americas, new and neglected viruses continue to emerge and spread around the world. Due to a lack of existing vaccines or specific therapeutics, little other than supportive care and attempts to interrupt transmission can be provided during initial outbreaks. This has prompted a shift in vaccine design and development to identify novel epitopes and mechanisms of protection that may offer a broader range of protection against groups or whole families of viruses. Receptor-binding domains and other motifs within viral envelope proteins represent one excellent opportunity to target communal epitopes shared by related viruses. Similarly, for viruses where envelope participates in driving viral egress from infected cells, shared epitopes need to be identified to guide the development of broadly protective antibodies and vaccines. Here, we discuss recent advances in our understanding of broadly protective humoral responses for emerging viruses.
Collapse
|
14
|
Mathys L, Balzarini J. The role of cellular oxidoreductases in viral entry and virus infection-associated oxidative stress: potential therapeutic applications. Expert Opin Ther Targets 2015; 20:123-43. [PMID: 26178644 DOI: 10.1517/14728222.2015.1068760] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Cellular oxidoreductases catalyze thiol/disulfide exchange reactions in susceptible proteins and contribute to the cellular defense against oxidative stress. Oxidoreductases and oxidative stress are also involved in viral infections. In this overview, different aspects of the role of cellular oxidoreductases and oxidative stress during viral infections are discussed from a chemotherapeutic viewpoint. AREAS COVERED Entry of enveloped viruses into their target cells is triggered by the interaction of viral envelope glycoproteins with cellular (co)receptor(s) and depends on obligatory conformational changes in these viral envelope glycoproteins and/or cellular receptors. For some viruses, these conformational changes are mediated by cell surface-associated cellular oxidoreductases, which mediate disulfide bridge reductions in viral envelope glycoprotein(s). Therefore, targeting these oxidoreductases using oxidoreductase inhibitors might yield an interesting strategy to block viral entry of these viruses. Furthermore, since viral infections are often associated with systemic oxidative stress, contributing to disease progression, the enhancement of the cellular antioxidant defense systems might have potential as an adjuvant antiviral strategy, slowing down disease progression. EXPERT OPINION Promising antiviral data were obtained for both strategies. However, potential pitfalls have also been identified for these strategies, indicating that it is important to carefully assess the benefits versus risks of these antiviral strategies.
Collapse
Affiliation(s)
- Leen Mathys
- a 1 Rega Institute for Medical Research, KU Leuven , Minderbroedersstraat 10 blok x - bus 1030, Leuven, Belgium
| | - Jan Balzarini
- b 2 Rega Institute for Medical Research, KU Leuven , Minderbroedersstraat 10 blok x - bus 1030, Leuven, Belgium +32 16 3 37352 ; +32 16 3 37340 ;
| |
Collapse
|
15
|
Abstract
The current Ebola virus disease (EVD) outbreak in West Africa is the largest with over 5100 deaths in four West African countries as of 14 November 2014. EVD has high case-fatality rates but no licensed treatment or vaccine is yet available. Several vaccine candidates that protected nonhuman primates are not yet available for clinical use. Slow development of vaccine-stimulated immunity, sporadic nature and fast progression of EVD underlines the need for the development of effective postexposure therapeutic drugs. WHO encouraged the use of untested drugs for EVD to curb the fast-spreading outbreak. Here, we summarize therapeutics for EVD including monoclonal antibody-based therapy and inhibitors of viral replication including our recently developed small-molecule inhibitors of VP30 dephosphorylation.
Collapse
Affiliation(s)
- Marina Jerebtsova
- Department of Microbiology, Howard University, Washington, DC 20059, USA
| | - Sergei Nekhai
- Department of Microbiology, Howard University, Washington, DC 20059, USA ; Center for Sickle Cell Disease, Howard University, Washington, DC 20059, USA ; Department of Medicine, Howard University, Washington, DC 20059, USA ; Department of Pharmacology, Howard University, Washington, DC 20059, USA
| |
Collapse
|
16
|
Neutralization of Virus Infectivity by Antibodies: Old Problems in New Perspectives. ACTA ACUST UNITED AC 2014; 2014. [PMID: 27099867 DOI: 10.1155/2014/157895] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Neutralizing antibodies (NAbs) can be both sufficient and necessary for protection against viral infections, although they sometimes act in concert with cellular immunity. Successful vaccines against viruses induce NAbs but vaccine candidates against some major viral pathogens, including HIV-1, have failed to induce potent and effective such responses. Theories of how antibodies neutralize virus infectivity have been formulated and experimentally tested since the 1930s; and controversies about the mechanistic and quantitative bases for neutralization have continually arisen. Soluble versions of native oligomeric viral proteins that mimic the functional targets of neutralizing antibodies now allow the measurement of the relevant affinities of NAbs. Thereby the neutralizing occupancies on virions can be estimated and related to the potency of the NAbs. Furthermore, the kinetics and stoichiometry of NAb binding can be compared with neutralizing efficacy. Recently, the fundamental discovery that the intracellular factor TRIM21 determines the degree of neutralization of adenovirus has provided new mechanistic and quantitative insights. Since TRIM21 resides in the cytoplasm, it would not affect the neutralization of enveloped viruses, but its range of activity against naked viruses will be important to uncover. These developments bring together the old problems of virus neutralization-mechanism, stoichiometry, kinetics, and efficacy-from surprising new angles.
Collapse
|