1
|
Shi YJ, Li JQ, Zhang HQ, Deng CL, Zhu QX, Zhang B, Li XD. A high throughput antiviral screening platform for alphaviruses based on Semliki Forest virus expressing eGFP reporter gene. Virol Sin 2023; 38:585-594. [PMID: 37390870 PMCID: PMC10436050 DOI: 10.1016/j.virs.2023.06.007] [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: 02/03/2023] [Accepted: 06/20/2023] [Indexed: 07/02/2023] Open
Abstract
Alphaviruses, which contain a variety of mosquito-borne pathogens, are important pathogens of emerging/re-emerging infectious diseases and potential biological weapons. Currently, no specific antiviral drugs are available for the treatment of alphaviruses infection. For most highly pathogenic alphaviruses are classified as risk group-3 agents, the requirement of biosafety level 3 (BSL-3) facilities limits the live virus-based antiviral study. To facilitate the antiviral development of alphaviruses, we developed a high throughput screening (HTS) platform based on a recombinant Semliki Forest virus (SFV) which can be manipulated in BSL-2 laboratory. Using the reverse genetics approach, the recombinant SFV and SFV reporter virus expressing eGFP (SFV-eGFP) were successfully rescued. The SFV-eGFP reporter virus exhibited robust eGFP expression and remained relatively stable after four passages in BHK-21 cells. Using a broad-spectrum alphavirus inhibitor ribavirin, we demonstrated that the SFV-eGFP can be used as an effective tool for antiviral study. The SFV-eGFP reporter virus-based HTS assay in a 96-well format was then established and optimized with a robust Z' score. A section of reference compounds that inhibit highly pathogenic alphaviruses were used to validate that the SFV-eGFP reporter virus-based HTS assay enables rapid screening of potent broad-spectrum inhibitors of alphaviruses. This assay provides a safe and convenient platform for antiviral study of alphaviruses.
Collapse
Affiliation(s)
- Yu-Jia Shi
- Hunan Normal University, School of Medicine, Changsha, 410081, China
| | - Jia-Qi Li
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hong-Qing Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Cheng-Lin Deng
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qin-Xuan Zhu
- Hunan Normal University, School of Medicine, Changsha, 410081, China
| | - Bo Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xiao-Dan Li
- Hunan Normal University, School of Medicine, Changsha, 410081, China.
| |
Collapse
|
2
|
Hu X, Morazzani E, Compton JR, Harmon M, Soloveva V, Glass PJ, Garcia AD, Marugan JJ, Legler PM. In Silico Screening of Inhibitors of the Venezuelan Equine Encephalitis Virus Nonstructural Protein 2 Cysteine Protease. Viruses 2023; 15:1503. [PMID: 37515189 PMCID: PMC10385868 DOI: 10.3390/v15071503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
The Venezuelan equine encephalitis virus (VEEV) nonstructural protein 2 (nsP2) cysteine protease (EC 3.4.22.B79) is essential for viral replication. High throughput in silico/in vitro screening using a focused set of known cysteine protease inhibitors identified two epoxysuccinyl prodrugs, E64d and CA074 methyl ester (CA074me) and a reversible oxindole inhibitor. Here, we determined the X-ray crystal structure of the CA074-inhibited nsP2 protease and compared it with our E64d-inhibited structure. We found that the two inhibitors occupy different locations in the protease. We designed hybrid inhibitors with improved potency. Virus yield reduction assays confirmed that the viral titer was reduced by >5 logs with CA074me. Cell-based assays showed reductions in viral replication for CHIKV, VEEV, and WEEV, and weaker inhibition of EEEV by the hybrid inhibitors. The most potent was NCGC00488909-01 which had an EC50 of 1.76 µM in VEEV-Trd-infected cells; the second most potent was NCGC00484087 with an EC50 = 7.90 µM. Other compounds from the NCATS libraries such as the H1 antihistamine oxatomide (>5-log reduction), emetine, amsacrine an intercalator (NCGC0015113), MLS003116111-01, NCGC00247785-13, and MLS00699295-01 were found to effectively reduce VEEV viral replication in plaque assays. Kinetic methods demonstrated time-dependent inhibition by the hybrid inhibitors of the protease with NCGC00488909-01 (Ki = 3 µM) and NCGC00484087 (Ki = 5 µM). Rates of inactivation by CA074 in the presence of 6 mM CaCl2, MnCl2, or MgCl2 were measured with varying concentrations of inhibitor, Mg2+ and Mn2+ slightly enhanced inhibitor binding (3 to 6-fold). CA074 inhibited not only the VEEV nsP2 protease but also that of CHIKV and WEEV.
Collapse
Affiliation(s)
- Xin Hu
- National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| | - Elaine Morazzani
- General Dynamics Information Technology, Falls Church, VA 22042, USA
| | - Jaimee R Compton
- Center for Bio/Molecular Science and Engineering (CBMSE), Naval Research Laboratory, Washington, DC 20375, USA
| | - Moeshia Harmon
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, MS 39217, USA
| | - Veronica Soloveva
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA
| | - Pamela J Glass
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA
| | - Andres Dulcey Garcia
- National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| | - Juan J Marugan
- National Center for Advancing Translational Sciences (NCATS), Rockville, MD 20850, USA
| | - Patricia M Legler
- Center for Bio/Molecular Science and Engineering (CBMSE), Naval Research Laboratory, Washington, DC 20375, USA
| |
Collapse
|
3
|
Cao X, Yang D, Parvathareddy J, Chu YK, Kim EJ, Fitz-Henley JN, Li X, Lukka PB, Parmar KR, Temrikar ZH, Dhole P, Adcock RS, Gabbard J, Bansal S, Lee J, Zalduondo L, Hayes E, Stabenow J, Meibohm B, Fitzpatrick EA, Bailey K, Campos RK, Julander JG, Rossi SL, Chung D, Jonsson CB, Golden JE. Efficacy of a brain-penetrant antiviral in lethal Venezuelan and eastern equine encephalitis mouse models. Sci Transl Med 2023; 15:eabl9344. [PMID: 37043558 PMCID: PMC11577637 DOI: 10.1126/scitranslmed.abl9344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 03/24/2023] [Indexed: 04/14/2023]
Abstract
Venezuelan and eastern equine encephalitis viruses (VEEV and EEEV, respectively) are mosquito-borne, neuroinvasive human pathogens for which no FDA-approved therapeutic exists. Besides the biothreat posed by these viruses when aerosolized, arthropod transmission presents serious health risks to humans, as demonstrated by the 2019 outbreak of EEE disease in the United States that resulted in 38 confirmed cases, 19 deaths, and neurological effects in survivors. Here, we describe the discovery of a 2-pyrrolidinoquinazolinone scaffold, efficiently synthesized in two to five steps, whose structural optimization resulted in profound antiviral activity. The lead quinazolinone, BDGR-49, potently reduced cellular VEEV and EEEV titers by >7 log at 1 μM and exhibited suitable intravenous and oral pharmacokinetic profiles in BALB/c mice to achieve excellent brain exposure. Outstanding in vivo efficacy was observed in several lethal, subcutaneous infection mouse models using an 8-day dosing regimen. Prophylactically administered BDGR-49 at 25 mg kg-1 per day fully protected against a 10× LD50 VEEV Trinidad donkey (TrD) challenge in BALB/c mice. Similarly, we observed 70% protection when 10× LD50 EEEV FL93-939-infected C57BL/6 mice were treated prophylactically with BDGR-49 at 50 mg kg-1 per day. Last, we observed 100% therapeutic efficacy when mice, challenged with 10× LD50 VEEV TrD, were dosed at 48 hours after infection with BDGR-49 at 25 mg kg-1 per day. Mouse brain viral titers at 96 hours after infection were reduced to values near the limit of detection. Collectively, these results underscore the substantial development potential of a well-tolerated, brain-penetrant lead compound that shows promise in preventing and treating encephalitic alphavirus disease.
Collapse
Affiliation(s)
- Xufeng Cao
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dong Yang
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jyothi Parvathareddy
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Yong-Kyu Chu
- Center for Predictive Medicine, Department of Microbiology Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Eun Jung Kim
- Center for Predictive Medicine, Department of Microbiology Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Jhewelle N Fitz-Henley
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Xiaoyu Li
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Pradeep B Lukka
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Keyur R Parmar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Zaid H Temrikar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Priya Dhole
- Center for Predictive Medicine, Department of Microbiology Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Robert Scott Adcock
- Center for Predictive Medicine, Department of Microbiology Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Jon Gabbard
- Center for Predictive Medicine, Department of Microbiology Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Shruti Bansal
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jasper Lee
- Departments of Microbiology, Immunology, Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Lillian Zalduondo
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ernestine Hayes
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jennifer Stabenow
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Elizabeth A Fitzpatrick
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Departments of Microbiology, Immunology, Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Kevin Bailey
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Rafael K Campos
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Justin G Julander
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Shannan L Rossi
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Donghoon Chung
- Center for Predictive Medicine, Department of Microbiology Immunology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Colleen B Jonsson
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Departments of Microbiology, Immunology, Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jennifer E Golden
- School of Pharmacy, Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
4
|
Ogorek TJ, Golden JE. Advances in the Development of Small Molecule Antivirals against Equine Encephalitic Viruses. Viruses 2023; 15:413. [PMID: 36851628 PMCID: PMC9958955 DOI: 10.3390/v15020413] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/29/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
Venezuelan, western, and eastern equine encephalitic alphaviruses (VEEV, WEEV, and EEEV, respectively) are arboviruses that are highly pathogenic to equines and cause significant harm to infected humans. Currently, human alphavirus infection and the resulting diseases caused by them are unmitigated due to the absence of approved vaccines or therapeutics for general use. These circumstances, combined with the unpredictability of outbreaks-as exemplified by a 2019 EEE surge in the United States that claimed 19 patient lives-emphasize the risks posed by these viruses, especially for aerosolized VEEV and EEEV which are potential biothreats. Herein, small molecule inhibitors of VEEV, WEEV, and EEEV are reviewed that have been identified or advanced in the last five years since a comprehensive review was last performed. We organize structures according to host- versus virus-targeted mechanisms, highlight cellular and animal data that are milestones in the development pipeline, and provide a perspective on key considerations for the progression of compounds at early and later stages of advancement.
Collapse
Affiliation(s)
- Tyler J. Ogorek
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jennifer E. Golden
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
5
|
Biselli R, Nisini R, Lista F, Autore A, Lastilla M, De Lorenzo G, Peragallo MS, Stroffolini T, D’Amelio R. A Historical Review of Military Medical Strategies for Fighting Infectious Diseases: From Battlefields to Global Health. Biomedicines 2022; 10:2050. [PMID: 36009598 PMCID: PMC9405556 DOI: 10.3390/biomedicines10082050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/17/2022] Open
Abstract
The environmental conditions generated by war and characterized by poverty, undernutrition, stress, difficult access to safe water and food as well as lack of environmental and personal hygiene favor the spread of many infectious diseases. Epidemic typhus, plague, malaria, cholera, typhoid fever, hepatitis, tetanus, and smallpox have nearly constantly accompanied wars, frequently deeply conditioning the outcome of battles/wars more than weapons and military strategy. At the end of the nineteenth century, with the birth of bacteriology, military medical researchers in Germany, the United Kingdom, and France were active in discovering the etiological agents of some diseases and in developing preventive vaccines. Emil von Behring, Ronald Ross and Charles Laveran, who were or served as military physicians, won the first, the second, and the seventh Nobel Prize for Physiology or Medicine for discovering passive anti-diphtheria/tetanus immunotherapy and for identifying mosquito Anopheline as a malaria vector and plasmodium as its etiological agent, respectively. Meanwhile, Major Walter Reed in the United States of America discovered the mosquito vector of yellow fever, thus paving the way for its prevention by vector control. In this work, the military relevance of some vaccine-preventable and non-vaccine-preventable infectious diseases, as well as of biological weapons, and the military contributions to their control will be described. Currently, the civil-military medical collaboration is getting closer and becoming interdependent, from research and development for the prevention of infectious diseases to disasters and emergencies management, as recently demonstrated in Ebola and Zika outbreaks and the COVID-19 pandemic, even with the high biocontainment aeromedical evacuation, in a sort of global health diplomacy.
Collapse
Affiliation(s)
- Roberto Biselli
- Ispettorato Generale della Sanità Militare, Stato Maggiore della Difesa, Via S. Stefano Rotondo 4, 00184 Roma, Italy
| | - Roberto Nisini
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Roma, Italy
| | - Florigio Lista
- Dipartimento Scientifico, Policlinico Militare, Comando Logistico dell’Esercito, Via S. Stefano Rotondo 4, 00184 Roma, Italy
| | - Alberto Autore
- Osservatorio Epidemiologico della Difesa, Ispettorato Generale della Sanità Militare, Stato Maggiore della Difesa, Via S. Stefano Rotondo 4, 00184 Roma, Italy
| | - Marco Lastilla
- Istituto di Medicina Aerospaziale, Comando Logistico dell’Aeronautica Militare, Viale Piero Gobetti 2, 00185 Roma, Italy
| | - Giuseppe De Lorenzo
- Comando Generale dell’Arma dei Carabinieri, Dipartimento per l’Organizzazione Sanitaria e Veterinaria, Viale Romania 45, 00197 Roma, Italy
| | - Mario Stefano Peragallo
- Centro Studi e Ricerche di Sanità e Veterinaria, Comando Logistico dell’Esercito, Via S. Stefano Rotondo 4, 00184 Roma, Italy
| | - Tommaso Stroffolini
- Dipartimento di Malattie Infettive e Tropicali, Policlinico Umberto I, 00161 Roma, Italy
| | - Raffaele D’Amelio
- Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Via di Grottarossa 1035-1039, 00189 Roma, Italy
| |
Collapse
|
6
|
Painter GR, Natchus MG, Cohen O, Holman W, Painter WP. Developing a direct acting, orally available antiviral agent in a pandemic: the evolution of molnupiravir as a potential treatment for COVID-19. Curr Opin Virol 2021; 50:17-22. [PMID: 34271264 PMCID: PMC8277160 DOI: 10.1016/j.coviro.2021.06.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 01/22/2023]
Abstract
Despite the availability of vaccines, there remains an urgent need for antiviral drugs with potent activity against SARS-CoV-2, the cause of COVID-19. Millions of people are immune-suppressed and may not be able to mount a fully protective immune response after vaccination. There is also an increasingly critical need for a drug to cover emerging SARS-CoV-2 variants, against which existing vaccines may be less effective. Here, we describe the evolution of molnupiravir (EIDD-2801, MK-4482), a broad-spectrum antiviral agent originally designed for the treatment of Alphavirus infections, into a potential drug for the prevention and treatment of COVID-19. When the pandemic began, molnupiravir was in pre-clinical development for the treatment of seasonal influenza. As COVID-19 spread, the timeline for the development program was moved forward significantly, and focus shifted to treatment of coronavirus infections. Real time consultation with regulatory authorities aided in making the acceleration of the program possible.
Collapse
Affiliation(s)
- George R Painter
- Emory Institute for Drug Development (EIDD), Emory University, Atlanta, GA, USA; Drug Innovation Ventures at Emory (DRIVE), Atlanta, GA, USA; Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Michael G Natchus
- Emory Institute for Drug Development (EIDD), Emory University, Atlanta, GA, USA
| | - Oren Cohen
- Covance Clinical Research Unit Ltd., Springfield House, Hyde Street, Leeds LS2 9LH, UK
| | - Wendy Holman
- Ridgeback Biotherapeutics LP, 3480 Main Highway, Unit 402, Miami, Florida 33133, USA
| | - Wendy P Painter
- Ridgeback Biotherapeutics LP, 3480 Main Highway, Unit 402, Miami, Florida 33133, USA
| |
Collapse
|
7
|
Barraza SJ, Sindac JA, Dobry CJ, Delekta PC, Lee PH, Miller DJ, Larsen SD. Synthesis and biological activity of conformationally restricted indole-based inhibitors of neurotropic alphavirus replication: Generation of a three-dimensional pharmacophore. Bioorg Med Chem Lett 2021; 46:128171. [PMID: 34098081 PMCID: PMC8272561 DOI: 10.1016/j.bmcl.2021.128171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/26/2021] [Accepted: 05/30/2021] [Indexed: 11/29/2022]
Abstract
We have previously reported the development of indole-based CNS-active antivirals for the treatment of neurotropic alphavirus infection, but further optimization is impeded by a lack of knowledge of the molecular target and binding site. Herein we describe the design, synthesis and evaluation of a series of conformationally restricted analogues with the dual objectives of improving potency/selectivity and identifying the most bioactive conformation. Although this campaign was only modestly successful at improving potency, the sharply defined SAR of the rigid analogs enabled the definition of a three-dimensional pharmacophore, which we believe will be of value in further analog design and virtual screening for alternative antiviral leads.
Collapse
Affiliation(s)
- Scott J Barraza
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, United States
| | - Janice A Sindac
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, United States
| | - Craig J Dobry
- Departments of Internal Medicine and Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Philip C Delekta
- Departments of Internal Medicine and Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Pil H Lee
- Vahlteich Medicinal Chemistry Core, University of Michigan, Ann Arbor, MI 48109, United States; Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, United States
| | - David J Miller
- Departments of Internal Medicine and Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Scott D Larsen
- Vahlteich Medicinal Chemistry Core, University of Michigan, Ann Arbor, MI 48109, United States; Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
8
|
Hasan SS, Dey D, Singh S, Martin M. The Structural Biology of Eastern Equine Encephalitis Virus, an Emerging Viral Threat. Pathogens 2021; 10:pathogens10080973. [PMID: 34451437 PMCID: PMC8400090 DOI: 10.3390/pathogens10080973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/21/2021] [Accepted: 07/28/2021] [Indexed: 11/16/2022] Open
Abstract
Alphaviruses are arboviruses that cause arthritis and encephalitis in humans. Eastern Equine Encephalitis Virus (EEEV) is a mosquito-transmitted alphavirus that is implicated in severe encephalitis in humans with high mortality. However, limited insights are available into the fundamental biology of EEEV and residue-level details of its interactions with host proteins. In recent years, outbreaks of EEEV have been reported mainly in the United States, raising concerns about public safety. This review article summarizes recent advances in the structural biology of EEEV based mainly on single-particle cryogenic electron microscopy (cryoEM) structures. Together with functional analyses of EEEV and related alphaviruses, these structural investigations provide clues to how EEEV interacts with host proteins, which may open avenues for the development of therapeutics.
Collapse
Affiliation(s)
- S. Saif Hasan
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore, MD 21201, USA; (D.D.); (S.S.); (M.M.)
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 9600 Gudelsky Drive, Rockville, MD 20850, USA
- University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland Medical Center, 22. S. Greene St., Baltimore, MD 21201, USA
- Correspondence:
| | - Debajit Dey
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore, MD 21201, USA; (D.D.); (S.S.); (M.M.)
| | - Suruchi Singh
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore, MD 21201, USA; (D.D.); (S.S.); (M.M.)
| | - Matthew Martin
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore, MD 21201, USA; (D.D.); (S.S.); (M.M.)
| |
Collapse
|
9
|
Shechter S, Thomas DR, Jans DA. Application of In Silico and HTS Approaches to Identify Nuclear Import Inhibitors for Venezuelan Equine Encephalitis Virus Capsid Protein: A Case Study. Front Chem 2020; 8:573121. [PMID: 33505952 PMCID: PMC7832173 DOI: 10.3389/fchem.2020.573121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/12/2020] [Indexed: 01/16/2023] Open
Abstract
The development of new drugs is costly and time-consuming, with estimates of over $US1 billion and 15 years for a product to reach the market. As understanding of the molecular basis of disease improves, various approaches have been used to target specific molecular interactions in the search for effective drugs. These include high-throughput screening (HTS) for novel drug identification and computer-aided drug design (CADD) to assess the properties of putative drugs before experimental work begins. We have applied conventional HTS and CADD approaches to the problem of identifying antiviral compounds to limit infection by Venezuelan equine encephalitis virus (VEEV). Nuclear targeting of the VEEV capsid (CP) protein through interaction with the host nuclear import machinery has been shown to be essential for viral pathogenicity, with viruses incapable of this interaction being greatly attenuated. Our previous conventional HTS and in silico structure-based drug design (SBDD) screens were successful in identifying novel inhibitors of CP interaction with the host nuclear import machinery, thus providing a unique opportunity to assess the relative value of the two screening approaches directly. This focused review compares and contrasts the two screening approaches, together with the properties of the inhibitors identified, as a case study for parallel use of the two approaches to identify antivirals. The utility of SBDD screens, especially when used in parallel with traditional HTS, in identifying agents of interest to target the host-pathogen interface is highlighted.
Collapse
Affiliation(s)
- Sharon Shechter
- Shechter Computational Solutions, Andover, MA, United States.,Department of Chemistry, College of Sciences, University of Massachusetts Lowell, Lowell, MA, United States
| | - David R Thomas
- Nuclear Signalling Laboratory, Department of Biochemistry and Molecular Biology, Biomedical Discovery Institute, Monash University, Monash, VIC, Australia
| | - David A Jans
- Nuclear Signalling Laboratory, Department of Biochemistry and Molecular Biology, Biomedical Discovery Institute, Monash University, Monash, VIC, Australia
| |
Collapse
|
10
|
Williamson LE, Gilliland T, Yadav PK, Binshtein E, Bombardi R, Kose N, Nargi RS, Sutton RE, Durie CL, Armstrong E, Carnahan RH, Walker LM, Kim AS, Fox JM, Diamond MS, Ohi MD, Klimstra WB, Crowe JE. Human Antibodies Protect against Aerosolized Eastern Equine Encephalitis Virus Infection. Cell 2020; 183:1884-1900.e23. [PMID: 33301709 DOI: 10.1016/j.cell.2020.11.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/23/2020] [Accepted: 11/06/2020] [Indexed: 12/19/2022]
Abstract
Eastern equine encephalitis virus (EEEV) is one of the most virulent viruses endemic to North America. No licensed vaccines or antiviral therapeutics are available to combat this infection, which has recently shown an increase in human cases. Here, we characterize human monoclonal antibodies (mAbs) isolated from a survivor of natural EEEV infection with potent (<20 pM) inhibitory activity of EEEV. Cryo-electron microscopy reconstructions of two highly neutralizing mAbs, EEEV-33 and EEEV-143, were solved in complex with chimeric Sindbis/EEEV virions to 7.2 Å and 8.3 Å, respectively. The mAbs recognize two distinct antigenic sites that are critical for inhibiting viral entry into cells. EEEV-33 and EEEV-143 protect against disease following stringent lethal aerosol challenge of mice with highly pathogenic EEEV. These studies provide insight into the molecular basis for the neutralizing human antibody response against EEEV and can facilitate development of vaccines and candidate antibody therapeutics.
Collapse
Affiliation(s)
- Lauren E Williamson
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN 37232, USA
| | - Theron Gilliland
- The Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 165261, USA
| | - Pramod K Yadav
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elad Binshtein
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robin Bombardi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nurgun Kose
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rachel S Nargi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rachel E Sutton
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Clarissa L Durie
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Erica Armstrong
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robert H Carnahan
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lauren M Walker
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN 37232, USA
| | - Arthur S Kim
- Department of Medicine, Washington University, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University, St. Louis, MO 63110, USA
| | - Julie M Fox
- Department of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Michael S Diamond
- Department of Medicine, Washington University, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University, St. Louis, MO 63110, USA
| | - Melanie D Ohi
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - William B Klimstra
- The Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 165261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 165261, USA
| | - James E Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN 37232, USA; The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
11
|
Zhang H, Harmon M, Radoshitzky SR, Soloveva V, Kane CD, Duplantier AJ, Ogungbe IV. Vinyl Sulfone-Based Inhibitors of Nonstructural Protein 2 Block the Replication of Venezuelan Equine Encephalitis Virus. ACS Med Chem Lett 2020; 11:2139-2145. [PMID: 33214821 DOI: 10.1021/acsmedchemlett.0c00215] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 09/02/2020] [Indexed: 01/01/2023] Open
Abstract
Emerging infectious diseases like those caused by arboviruses such as Venezuelan equine encephalitis virus (VEEV) pose a serious threat to public health systems. Development of medical countermeasures against emerging infectious diseases are of utmost importance. In this work, an acrylate and vinyl sulfone-based chemical series was investigated as promising starting scaffolds against VEEV and as inhibitors of the cysteine protease domain of VEEV's nonstructural protein 2 (nsP2). Primary screen and dose response studies were performed to evaluate the potency and cytotoxicity of the compounds. The results provide structural insights into a new class of potent nonpeptidic covalent inhibitors of nsP2 cysteine protease represented by compound 11 (VEEV TrD, EC50 = 2.4 μM (HeLa), 1.6 μM (Vero E6)). These results may facilitate the evolution of the compounds into selective and broad-spectrum anti-alphaviral drug leads.
Collapse
Affiliation(s)
- Huaisheng Zhang
- Department of Chemistry, Physics, and Atmospheric Sciences, Jackson State University, Jackson, Mississippi 39217-0095, United States
| | - Moeshia Harmon
- Department of Chemistry, Physics, and Atmospheric Sciences, Jackson State University, Jackson, Mississippi 39217-0095, United States
| | - Sheli R. Radoshitzky
- The Geneva Foundation, Countermeasure Development Division, U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland 21702-5011, United States
| | - Veronica Soloveva
- Cherokee Nation Assurance, Countermeasure Development Division, U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland 21702-5011, United States
| | - Christopher D. Kane
- Research Program Office, U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland 21702-5011, United States
| | - Allen J. Duplantier
- Cherokee Nation Assurance, Countermeasure Development Division, U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland 21702-5011, United States
| | - Ifedayo Victor Ogungbe
- Department of Chemistry, Physics, and Atmospheric Sciences, Jackson State University, Jackson, Mississippi 39217-0095, United States
| |
Collapse
|
12
|
Painter GR, Bowen RA, Bluemling GR, DeBergh J, Edpuganti V, Gruddanti PR, Guthrie DB, Hager M, Kuiper DL, Lockwood MA, Mitchell DG, Natchus MG, Sticher ZM, Kolykhalov AA. The prophylactic and therapeutic activity of a broadly active ribonucleoside analog in a murine model of intranasal venezuelan equine encephalitis virus infection. Antiviral Res 2019; 171:104597. [DOI: 10.1016/j.antiviral.2019.104597] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 11/28/2022]
|
13
|
Bhat SM, Mudgal PP, N S, Arunkumar G. Spectrum of candidate molecules against Chikungunya virus - an insight into the antiviral screening platforms. Expert Rev Anti Infect Ther 2019; 17:243-264. [PMID: 30889372 DOI: 10.1080/14787210.2019.1595591] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Chikungunya disease has undergone a phenomenal transition in its status from being recognized as a sporadic infection to acquiring a global prominence over the last couple of decades. The causative agent behind the explosive epidemics worldwide is the re-emerging pathogen, Chikungunya virus (CHIKV). Areas covered: The current review discusses all the possible avenues of antiviral research towards combating CHIKV infection. Aspects of antiviral drug discovery such as antiviral targets, candidate molecules screened, and the various criteria to be a potential inhibitor are all discussed at length. Existing antiviral drug screening tools for CHIKV and their applications are thoroughly described. Clinical trial status of agents with therapeutic potential has been updated with special mention of candidate molecules under patent approval. Databases such as PubMed, Google Scholar, ScienceDirect, Google Patent, and Clinical Trial Registry platforms were referred. Expert opinion: The massive outbreaks of Chikungunya viral disease in the recent past and the serious health concerns imposed thereby, have driven the search for effective therapeutics. The greatest challenge being the non-availability of robust, reproducible, cost-effective and biologically accurate assay models. Nevertheless, there is a need to identify good models mimicking the appropriate microenvironment of an infectious setting.
Collapse
Affiliation(s)
- Shree Madhu Bhat
- a Manipal Centre for Virus Research , Manipal Academy of Higher Education (Deemed to be University) , Manipal , Karnataka , India
| | - Piya Paul Mudgal
- a Manipal Centre for Virus Research , Manipal Academy of Higher Education (Deemed to be University) , Manipal , Karnataka , India
| | - Sudheesh N
- a Manipal Centre for Virus Research , Manipal Academy of Higher Education (Deemed to be University) , Manipal , Karnataka , India
| | - Govindakarnavar Arunkumar
- a Manipal Centre for Virus Research , Manipal Academy of Higher Education (Deemed to be University) , Manipal , Karnataka , India
| |
Collapse
|
14
|
Lundberg L, Fontenot J, Lin SC, Pinkham C, Carey BD, Campbell CE, Kehn-Hall K. Venezuelan Equine Encephalitis Virus Capsid Implicated in Infection-Induced Cell Cycle Delay in vitro. Front Microbiol 2018; 9:3126. [PMID: 30631316 PMCID: PMC6315117 DOI: 10.3389/fmicb.2018.03126] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 12/03/2018] [Indexed: 02/06/2023] Open
Abstract
Venezuelan equine encephalitis virus (VEEV) is a positive sense, single-stranded RNA virus and member of the New World alphaviruses. It causes a biphasic febrile illness that can be accompanied by central nervous system involvement and moderate morbidity in humans and severe mortality in equines. The virus has a history of weaponization, lacks FDA-approved therapeutics and vaccines in humans, and is considered a select agent. Like other RNA viruses, VEEV replicates in the cytoplasm of infected cells and eventually induces apoptosis. The capsid protein, which contains a nuclear localization and a nuclear export sequence, induces a shutdown of host transcription and nucleocytoplasmic trafficking. Here we show that infection with VEEV causes a dysregulation of cell cycling and a delay in the G0/G1 phase in Vero cells and U87MG astrocytes. Cells infected with VEEV encoding a capsid NLS mutant or treated with the capsid-importin α interaction inhibitor G281-1485 were partially rescued from this cell cycle dysregulation. Pathway analysis of previously published RNA-sequencing data from VEEV infected U87MG astrocytes identified alterations of canonical pathways involving cell cycle, checkpoint regulation, and proliferation. Multiple cyclins including cyclin D1, cyclin A2 and cyclin E2 and other regulators of the cell cycle were downregulated in infected cells in a capsid NLS dependent manner. Loss of Rb phosphorylation, which is a substrate for cyclin/cdk complexes was also observed. These data demonstrate the importance of capsid nuclear localization and/or importin α binding for inducing cell cycle arrest and transcriptional downregulation of key cell cycle regulators.
Collapse
Affiliation(s)
- Lindsay Lundberg
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, United States
| | - Jacque Fontenot
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, United States
| | - Shih-Chao Lin
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, United States
| | - Chelsea Pinkham
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, United States
| | - Brian D Carey
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, United States
| | | | - Kylene Kehn-Hall
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, United States
| |
Collapse
|
15
|
Lundberg L, Brahms A, Hooper I, Carey B, Lin SC, Dahal B, Narayanan A, Kehn-Hall K. Repurposed FDA-Approved drug sorafenib reduces replication of Venezuelan equine encephalitis virus and other alphaviruses. Antiviral Res 2018; 157:57-67. [PMID: 29981794 DOI: 10.1016/j.antiviral.2018.07.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/30/2018] [Accepted: 07/02/2018] [Indexed: 12/12/2022]
Abstract
The New World alphaviruses -Venezuelan, eastern, and western equine encephalitis viruses (VEEV, EEEV, and WEEV respectively) - cause a febrile disease that is often lethal in equines and children and leads to long-term neurological sequelae in survivors. Endemic to the Americas, epizootic outbreaks of the three viruses occur sporadically in the continental United States. All three viruses aerosolize readily, replicate to high titers in cell culture, and have low infectious doses. Additionally, there are no FDA-approved vaccines or therapeutics for human use. To address the therapeutic gap, a high throughput assay utilizing a luciferase reporter virus, TC83-luc, was performed to screen a library of commercially available, FDA-approved drugs for antiviral activity. From a group of twenty compounds found to significantly decrease luminescence, the carcinoma therapeutic sorafenib inhibited replication of VEEV-TC83 and TrD in vitro. Additionally, sorafenib inhibited replication of EEEV and two Old World alphaviruses, Sindbis virus and chikungunya virus, at 8 and 16 h post-infection. Sorafenib caused no toxicity in Vero cells, and coupled with a low EC50 value, yielded a selectivity index of >19. Mechanism of actions studies suggest that sorafenib inhibited viral translation through dephosphorylation of several key proteins, including eIF4E and p70S6K, leading to a reduction in viral protein production and overall viral replication.
Collapse
Affiliation(s)
- Lindsay Lundberg
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Ashwini Brahms
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Idris Hooper
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Brian Carey
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Shih-Chao Lin
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Bibha Dahal
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Aarthi Narayanan
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Kylene Kehn-Hall
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA.
| |
Collapse
|
16
|
Burke CW, Froude JW, Miethe S, Hülseweh B, Hust M, Glass PJ. Human-Like Neutralizing Antibodies Protect Mice from Aerosol Exposure with Western Equine Encephalitis Virus. Viruses 2018; 10:v10040147. [PMID: 29587363 PMCID: PMC5923441 DOI: 10.3390/v10040147] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/14/2018] [Accepted: 03/22/2018] [Indexed: 02/07/2023] Open
Abstract
Western equine encephalitis virus (WEEV) causes symptoms in humans ranging from mild febrile illness to life-threatening encephalitis, and no human medical countermeasures are licensed. A previous study demonstrated that immune serum from vaccinated mice protected against lethal WEEV infection, suggesting the utility of antibodies for pre- and post-exposure treatment. Here, three neutralizing and one binding human-like monoclonal antibodies were evaluated against WEEV aerosol challenge. Dose-dependent protection was observed with two antibodies administered individually, ToR69-3A2 and ToR68-2C3. In vitro neutralization was not a critical factor for protection in this murine model, as ToR69-3A2 is a strong neutralizing antibody, and ToR68-2C3 is a non-neutralizing antibody. This result highlights the importance of both neutralizing and non-neutralizing antibodies in the protection of mice from WEEV lethality.
Collapse
MESH Headings
- Aerosols
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/immunology
- Antibodies, Neutralizing/administration & dosage
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/administration & dosage
- Antibodies, Viral/immunology
- Disease Models, Animal
- Encephalitis Virus, Western Equine/immunology
- Encephalomyelitis, Equine/immunology
- Encephalomyelitis, Equine/mortality
- Encephalomyelitis, Equine/prevention & control
- Encephalomyelitis, Equine/virology
- Immunization
- Mice
- Morbidity
- Mortality
Collapse
Affiliation(s)
- Crystal W Burke
- United States Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, USA.
| | - Jeffrey W Froude
- United States Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, USA.
| | - Sebastian Miethe
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr.7, 38106 Braunschweig, Germany.
| | - Birgit Hülseweh
- Wehrwissenschaftliches Institut für Schutztechnologien (WIS)-ABC-Schutz, Humboldtstr. 1, 29623 Munster, Germany.
| | - Michael Hust
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr.7, 38106 Braunschweig, Germany.
- YUMAB GmbH, Science Campus Braunschweig Süd, Inhoffenstr.7, 38124 Braunschweig, Germany.
| | - Pamela J Glass
- United States Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, USA.
| |
Collapse
|
17
|
Brown RS, Wan JJ, Kielian M. The Alphavirus Exit Pathway: What We Know and What We Wish We Knew. Viruses 2018; 10:E89. [PMID: 29470397 PMCID: PMC5850396 DOI: 10.3390/v10020089] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 02/19/2018] [Accepted: 02/20/2018] [Indexed: 12/28/2022] Open
Abstract
Alphaviruses are enveloped positive sense RNA viruses and include serious human pathogens, such as the encephalitic alphaviruses and Chikungunya virus. Alphaviruses are transmitted to humans primarily by mosquito vectors and include species that are classified as emerging pathogens. Alphaviruses assemble highly organized, spherical particles that bud from the plasma membrane. In this review, we discuss what is known about the alphavirus exit pathway during a cellular infection. We describe the viral protein interactions that are critical for virus assembly/budding and the host factors that are involved, and we highlight the recent discovery of cell-to-cell transmission of alphavirus particles via intercellular extensions. Lastly, we discuss outstanding questions in the alphavirus exit pathway that may provide important avenues for future research.
Collapse
Affiliation(s)
- Rebecca S Brown
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Judy J Wan
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Margaret Kielian
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
18
|
Identification of novel antivirals inhibiting recognition of Venezuelan equine encephalitis virus capsid protein by the Importin α/β1 heterodimer through high-throughput screening. Antiviral Res 2018; 151:8-19. [PMID: 29337164 DOI: 10.1016/j.antiviral.2018.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 01/08/2018] [Accepted: 01/08/2018] [Indexed: 11/24/2022]
Abstract
Although the alphavirus Venezuelan equine encephalitis virus (VEEV) has been the cause of multiple outbreaks resulting in extensive human and equine mortality and morbidity, there are currently no anti-VEEV therapeutics available. VEEV pathogenicity is largely dependent on targeting of the viral capsid protein (CP) to the host cell nucleus through the nuclear transporting importin (Imp) α/β1 heterodimer. Here we perform a high-throughput screen, combined with nested counterscreens to identify small molecules able to inhibit the Impα/β1:CP interaction for the first time. Several compounds were able to significantly reduce viral replication in infected cells. Compound G281-1564 in particular could inhibit VEEV replication at low μM concentration, while showing minimal toxicity, with steady state and dynamic quantitative microscopic measurements confirming its ability to inhibit CP nuclear import. This study establishes the principle that inhibitors of CP nucleocytoplasmic trafficking can have potent antiviral activity against VEEV, and represents a platform for future development of safe anti-VEEV compounds with high efficacy and specificity.
Collapse
|
19
|
Compton JR, Mickey MJ, Hu X, Marugan JJ, Legler PM. Mutation of Asn-475 in the Venezuelan Equine Encephalitis Virus nsP2 Cysteine Protease Leads to a Self-Inhibited State. Biochemistry 2017; 56:6221-6230. [PMID: 29064679 DOI: 10.1021/acs.biochem.7b00746] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The alphaviral nsP2 cysteine protease of the Venezuelan equine encephalitis virus (VEEV) is a validated antiviral drug target. Clan CN proteases contain a cysteine protease domain that is intimately packed with an S-adenosyl-l-methionine-dependent RNA methyltransferase (SAM MTase) domain. Within a cleft formed at the interface of these two domains, the peptide substrate is thought to bind. The nucleophilic cysteine can be found within a conserved motif, 475NVCWAK480, which differs from that of papain (22CGSCWAFS29). Mutation of the motif residue, N475, to alanine unexpectedly produced a self-inhibited state in which the N-terminal residues flipped into the substrate-binding cleft. Notably, the N-terminal segment was not hydrolyzed-consistent with a catalytically incompetent state. The N475A mutation resulted in a 70-fold decrease in kcat/Km. A side chain-substrate interaction was predicted by the structure; the S701A mutation led to a 17-fold increase in Km. An Asn at the n-2 position relative to the Cys was also found in the coronaviral papain-like proteases/deubiquitinases (PLpro) of the SARS and MERS viruses, and in several papain-like human ubiquitin specific proteases (USP). The large conformational change in the N475A variant suggests that Asn-475 plays an important role in stabilizing the N-terminal residues and in orienting the carbonyl during nucleophilic attack but does not directly hydrogen bond the oxyanion. The state trapped in crystallo is an unusual result of site-directed mutagenesis but reveals the role of this highly conserved Asn and identifies key substrate-binding contacts that may be exploited by peptide-like inhibitors.
Collapse
Affiliation(s)
- Jaimee R Compton
- U.S. Naval Research Laboratory , 4555 Overlook Avenue, Washington, D.C. 20375, United States
| | | | - Xin Hu
- National Center for Advancing Translational Sciences, National Institutes of Health , Rockville, Maryland 20850, United States
| | - Juan J Marugan
- National Center for Advancing Translational Sciences, National Institutes of Health , Rockville, Maryland 20850, United States
| | - Patricia M Legler
- U.S. Naval Research Laboratory , 4555 Overlook Avenue, Washington, D.C. 20375, United States
| |
Collapse
|
20
|
Gollapudi D, Wycuff DL, Schwartz RM, Cooper JW, Cheng KC. Development of high‐throughput and high sensitivity capillary gel electrophoresis platform method for Western, Eastern, and Venezuelan equine encephalitis (WEVEE) virus like particles (VLPs) purity determination and characterization. Electrophoresis 2017; 38:2610-2621. [DOI: 10.1002/elps.201700217] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/25/2017] [Accepted: 06/25/2017] [Indexed: 12/11/2022]
Affiliation(s)
| | - Diane L Wycuff
- Vaccine Production Program VRC NIAID NIH Gaithersburg MD USA
| | - Richard M Schwartz
- Vaccine Production Program VRC NIAID NIH Gaithersburg MD USA
- Synlogic Cambridge MA USA
| | | | - KC Cheng
- Vaccine Production Program VRC NIAID NIH Gaithersburg MD USA
| |
Collapse
|
21
|
Porter AI, Erwin-Cohen RA, Twenhafel N, Chance T, Yee SB, Kern SJ, Norwood D, Hartman LJ, Parker MD, Glass PJ, DaSilva L. Characterization and pathogenesis of aerosolized eastern equine encephalitis in the common marmoset (Callithrix jacchus). Virol J 2017; 14:25. [PMID: 28173871 PMCID: PMC5297202 DOI: 10.1186/s12985-017-0687-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 01/18/2017] [Indexed: 02/02/2023] Open
Abstract
Background Licensed antiviral therapeutics and vaccines to protect against eastern equine encephalitis virus (EEEV) in humans currently do not exist. Animal models that faithfully recapitulate the clinical characteristics of human EEEV encephalitic disease, including fever, drowsiness, anorexia, and neurological signs such as seizures, are needed to satisfy requirements of the Food and Drug Administration (FDA) for clinical product licensing under the Animal Rule. Methods In an effort to meet this requirement, we estimated the median lethal dose and described the pathogenesis of aerosolized EEEV in the common marmoset (Callithrix jacchus). Five marmosets were exposed to aerosolized EEEV FL93-939 in doses ranging from 2.4 × 101 PFU to 7.95 × 105 PFU. Results The median lethal dose was estimated to be 2.05 × 102 PFU. Lethality was observed as early as day 4 post-exposure in the highest-dosed marmoset but animals at lower inhaled doses had a protracted disease course where humane study endpoint was not met until as late as day 19 post-exposure. Clinical signs were observed as early as 3 to 4 days post-exposure, including fever, ruffled fur, decreased grooming, and leukocytosis. Clinical signs increased in severity as disease progressed to include decreased body weight, subdued behavior, tremors, and lack of balance. Fever was observed as early as day 2–3 post-exposure in the highest dose groups and hypothermia was observed in several cases as animals became moribund. Infectious virus was found in several key tissues, including brain, liver, kidney, and several lymph nodes. Clinical hematology results included early neutrophilia, lymphopenia, and thrombocytopenia. Key pathological changes included meningoencephalitis and retinitis. Immunohistochemical staining for viral antigen was positive in the brain, retina, and lymph nodes. More intense and widespread IHC labeling occurred with increased aerosol dose. Conclusion We have estimated the medial lethal dose of aerosolized EEEV and described the pathology of clinical disease in the marmoset model. The results demonstrate that the marmoset is an animal model suitable for emulation of human EEEV disease in the development of medical countermeasures. Electronic supplementary material The online version of this article (doi:10.1186/s12985-017-0687-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aimee I Porter
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Virology Division, Frederick, MD, 21702, USA
| | - Rebecca A Erwin-Cohen
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Virology Division, Frederick, MD, 21702, USA.
| | | | | | - Steven B Yee
- Center for Aerobiological Sciences, Frederick, MD, 21702, USA
| | - Steven J Kern
- Research Support Division, Frederick, MD, 21702, USA
| | - David Norwood
- Diagnostic Systems Division, Frederick, MD, 21702, USA
| | | | - Michael D Parker
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Virology Division, Frederick, MD, 21702, USA
| | - Pamela J Glass
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Virology Division, Frederick, MD, 21702, USA
| | - Luis DaSilva
- Center for Aerobiological Sciences, Frederick, MD, 21702, USA
| |
Collapse
|
22
|
Zhang Y, Liu Q, Zhou B, Wang X, Chen S, Wang S. Ultra-sensitive chemiluminescence imaging DNA hybridization method in the detection of mosquito-borne viruses and parasites. Parasit Vectors 2017; 10:44. [PMID: 28122637 PMCID: PMC5267376 DOI: 10.1186/s13071-017-1975-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 01/07/2017] [Indexed: 11/10/2022] Open
Abstract
Background Mosquito-borne viruses (MBVs) and parasites (MBPs) are transmitted through hematophagous arthropods-mosquitoes to homoiothermous vertebrates. This study aims at developing a detection method to monitor the spread of mosquito-borne diseases to new areas and diagnose the infections caused by MBVs and MBPs. Methods In this assay, an ultra-sensitive chemiluminescence (CL) detection method was developed and used to simultaneously detect 19 common MBVs and MBPs. In vitro transcript RNA, virus-like particles (VLPs), and plasmids were established as positive or limit of detection (LOD) reference materials. Results MBVs and MBPs could be genotyped with high sensitivity and specificity. The cut-off values of probes were calculated. The absolute LODs of this strategy to detect serially diluted in vitro transcribed RNAs of MBVs and serially diluted plasmids of MBPs were 102–103copies/μl and 101–102copies/μl, respectively. Further, the LOD of detecting a strain of pre-quantified JEV was 101.8–100.8PFU/ml, fitted well in a linear regression model (coefficient of determination = 0.9678). Conclusions Ultra-sensitive CL imaging DNA hybridization was developed and could simultaneously detect various MBVs and MBPs. The method described here has the potential to provide considerable labor savings due to its ability to screen for 19 mosquito-borne pathogens simultaneously. Electronic supplementary material The online version of this article (doi:10.1186/s13071-017-1975-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yingjie Zhang
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.,Postdoctoral Research Workstation, 210th Hospital of the Chinese People's Liberation Army, Dalian, 116021, People's Republic of China
| | - Qiqi Liu
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.,Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious Diseases, Beijing, 100850, People's Republic of China
| | - Biao Zhou
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.,Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious Diseases, Beijing, 100850, People's Republic of China
| | - Xiaobo Wang
- Postdoctoral Research Workstation, 210th Hospital of the Chinese People's Liberation Army, Dalian, 116021, People's Republic of China
| | - Suhong Chen
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China. .,Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious Diseases, Beijing, 100850, People's Republic of China.
| | - Shengqi Wang
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China. .,Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious Diseases, Beijing, 100850, People's Republic of China.
| |
Collapse
|
23
|
Erwin-Cohen RA, Porter AI, Pittman PR, Rossi CA, DaSilva L. Human transcriptome response to immunization with live-attenuated Venezuelan equine encephalitis virus vaccine (TC-83): Analysis of whole blood. Hum Vaccin Immunother 2016; 13:169-179. [PMID: 27870591 PMCID: PMC5287313 DOI: 10.1080/21645515.2016.1227900] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Venezuelan equine encephalitis virus (VEEV) is an important human and animal alphavirus pathogen transmitted by mosquitoes. The virus is endemic in Central and South America, but has also caused equine outbreaks in southwestern areas of the United States. In an effort to better understand the molecular mechanisms of the development of immunity to this important pathogen, we performed transcriptional analysis from whole, unfractionated human blood of patients who had been immunized with the live-attenuated vaccine strain of VEEV, TC-83. We compared changes in the transcriptome between naïve individuals who were mock vaccinated with saline to responses of individuals who received TC-83. Significant transcriptional changes were noted at days 2, 7, and 14 following vaccination. The top canonical pathways revealed at early and intermediate time points (days 2 and 7) included the involvement of the classic interferon response, interferon-response factors, activation of pattern recognition receptors, and engagement of the inflammasome. By day 14, the top canonical pathways included oxidative phosphorylation, the protein ubiquitination pathway, natural killer cell signaling, and B-cell development. Biomarkers were identified that differentiate between vaccinees and control subjects, at early, intermediate, and late stages of the development of immunity as well as markers which were common to all 3 stages following vaccination but distinct from the sham-vaccinated control subjects. The study represents a novel examination of molecular processes that lead to the development of immunity against VEEV in humans and which may be of value as diagnostic targets, to enhance modern vaccine design, or molecular correlates of protection.
Collapse
Affiliation(s)
- Rebecca A Erwin-Cohen
- a Virology Division, United States Army Military Research Institute of Infectious Diseases (USAMRIID) , Frederick , MD , USA
| | - Aimee I Porter
- a Virology Division, United States Army Military Research Institute of Infectious Diseases (USAMRIID) , Frederick , MD , USA
| | - Phillip R Pittman
- b Division of Medicine, United States Army Military Research Institute of Infectious Diseases (USAMRIID) , Frederick , MD , USA
| | - Cynthia A Rossi
- c Diagnostics Systems Division, United States Army Military Research Institute of Infectious Diseases (USAMRIID) , Frederick , MD , USA
| | - Luis DaSilva
- d Center for Aerobiological Sciences, United States Army Military Research Institute of Infectious Diseases (USAMRIID) , Frederick , MD , USA
| |
Collapse
|
24
|
Lundberg L, Pinkham C, de la Fuente C, Brahms A, Shafagati N, Wagstaff KM, Jans DA, Tamir S, Kehn-Hall K. Selective Inhibitor of Nuclear Export (SINE) Compounds Alter New World Alphavirus Capsid Localization and Reduce Viral Replication in Mammalian Cells. PLoS Negl Trop Dis 2016; 10:e0005122. [PMID: 27902702 PMCID: PMC5130180 DOI: 10.1371/journal.pntd.0005122] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 10/21/2016] [Indexed: 01/28/2023] Open
Abstract
The capsid structural protein of the New World alphavirus, Venezuelan equine encephalitis virus (VEEV), interacts with the host nuclear transport proteins importin α/β1 and CRM1. Novel selective inhibitor of nuclear export (SINE) compounds, KPT-185, KPT-335 (verdinexor), and KPT-350, target the host's primary nuclear export protein, CRM1, in a manner similar to the archetypical inhibitor Leptomycin B. One major limitation of Leptomycin B is its irreversible binding to CRM1; which SINE compounds alleviate because they are slowly reversible. Chemically inhibiting CRM1 with these compounds enhanced capsid localization to the nucleus compared to the inactive compound KPT-301, as indicated by immunofluorescent confocal microscopy. Differences in extracellular versus intracellular viral RNA, as well as decreased capsid in cell free supernatants, indicated the inhibitors affected viral assembly, which led to a decrease in viral titers. The decrease in viral replication was confirmed using a luciferase-tagged virus and through plaque assays. SINE compounds had no effect on VEEV TC83_Cm, which encodes a mutated form of capsid that is unable to enter the nucleus. Serially passaging VEEV in the presence of KPT-185 resulted in mutations within the nuclear localization and nuclear export signals of capsid. Finally, SINE compound treatment also reduced the viral titers of the related eastern and western equine encephalitis viruses, suggesting that CRM1 maintains a common interaction with capsid proteins across the New World alphavirus genus.
Collapse
Affiliation(s)
- Lindsay Lundberg
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | - Chelsea Pinkham
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | - Cynthia de la Fuente
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | - Ashwini Brahms
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | - Nazly Shafagati
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| | - Kylie M. Wagstaff
- Nuclear Signaling Laboratory, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - David A. Jans
- Nuclear Signaling Laboratory, Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Sharon Tamir
- Karyopharm Therapeutics Inc., Massachusetts, United States of America
| | - Kylene Kehn-Hall
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, Virginia, United States of America
| |
Collapse
|
25
|
Campos-Gomez J, Ahmad F, Rodriguez E, Saeed MF. A novel cell-based assay to measure activity of Venezuelan equine encephalitis virus nsP2 protease. Virology 2016; 496:77-89. [PMID: 27261892 DOI: 10.1016/j.virol.2016.05.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 04/06/2016] [Accepted: 05/16/2016] [Indexed: 11/26/2022]
Abstract
The encephalitic alphaviruses encode nsP2 protease (nsP2pro), which because of its vital role in virus replication, represents an attractive target for therapeutic intervention. To facilitate the discovery of nsP2 inhibitors we have developed a novel assay for quantitative measurement of nsP2pro activity in a cell-based format. The assay is based on a substrate fusion protein consisting of eGFP and Gaussia luciferase (Gluc) linked together by a small peptide containing a VEEV nsp2pro cleavage sequence. The expression of the substrate protein in cells along with recombinant nsP2pro results in cleavage of the substrate protein resulting in extracellular release of free Gluc. The Gluc activity in supernatants corresponds to intracellular nsP2pro-mediated substrate cleavage; thus, providing a simple and convenient way to quantify nsP2pro activity. Here, we demonstrate potential utility of the assay in identification of nsP2pro inhibitors, as well as in investigations related to molecular characterization of nsP2pro.
Collapse
Affiliation(s)
- Javier Campos-Gomez
- Department of Infectious Diseases, Drug Discovery Division, Southern Research, Birmingham, AL 35205, United States
| | - Fahim Ahmad
- Department of Infectious Diseases, Drug Discovery Division, Southern Research, Birmingham, AL 35205, United States
| | - Efrain Rodriguez
- Department of Infectious Diseases, Drug Discovery Division, Southern Research, Birmingham, AL 35205, United States
| | - Mohammad F Saeed
- Department of Infectious Diseases, Drug Discovery Division, Southern Research, Birmingham, AL 35205, United States.
| |
Collapse
|
26
|
Venezuelan Equine Encephalitis Virus Induces Apoptosis through the Unfolded Protein Response Activation of EGR1. J Virol 2016; 90:3558-72. [PMID: 26792742 PMCID: PMC4794670 DOI: 10.1128/jvi.02827-15] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/08/2016] [Indexed: 12/11/2022] Open
Abstract
Venezuelan equine encephalitis virus (VEEV) is a previously weaponized arthropod-borne virus responsible for causing acute and fatal encephalitis in animal and human hosts. The increased circulation and spread in the Americas of VEEV and other encephalitic arboviruses, such as eastern equine encephalitis virus and West Nile virus, underscore the need for research aimed at characterizing the pathogenesis of viral encephalomyelitis for the development of novel medical countermeasures. The host-pathogen dynamics of VEEV Trinidad donkey-infected human astrocytoma U87MG cells were determined by carrying out RNA sequencing (RNA-Seq) of poly(A) and mRNAs. To identify the critical alterations that take place in the host transcriptome following VEEV infection, samples were collected at 4, 8, and 16 h postinfection and RNA-Seq data were acquired using an Ion Torrent PGM platform. Differential expression of interferon response, stress response factors, and components of the unfolded protein response (UPR) was observed. The protein kinase RNA-like endoplasmic reticulum kinase (PERK) arm of the UPR was activated, as the expression of both activating transcription factor 4 (ATF4) and CHOP (DDIT3), critical regulators of the pathway, was altered after infection. Expression of the transcription factor early growth response 1 (EGR1) was induced in a PERK-dependent manner. EGR1−/− mouse embryonic fibroblasts (MEFs) demonstrated lower susceptibility to VEEV-induced cell death than isogenic wild-type MEFs, indicating that EGR1 modulates proapoptotic pathways following VEEV infection. The influence of EGR1 is of great importance, as neuronal damage can lead to long-term sequelae in individuals who have survived VEEV infection. IMPORTANCE Alphaviruses represent a group of clinically relevant viruses transmitted by mosquitoes to humans. In severe cases, viral spread targets neuronal tissue, resulting in significant and life-threatening inflammation dependent on a combination of virus-host interactions. Currently there are no therapeutics for infections cause by encephalitic alphaviruses due to an incomplete understanding of their molecular pathogenesis. Venezuelan equine encephalitis virus (VEEV) is an alphavirus that is prevalent in the Americas and that is capable of infecting horses and humans. Here we utilized next-generation RNA sequencing to identify differential alterations in VEEV-infected astrocytes. Our results indicated that the abundance of transcripts associated with the interferon and the unfolded protein response pathways was altered following infection and demonstrated that early growth response 1 (EGR1) contributed to VEEV-induced cell death.
Collapse
|
27
|
Wu JQH. Virulence determinants of New World alphaviruses and broad-acting therapeutic strategies. Future Virol 2015. [DOI: 10.2217/fvl.15.8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
ABSTRACT New World alphaviruses of eastern equine encephalitis virus (EEEV), western equine encephalitis virus (WEEV) and Venezuelan equine encephalitis virus (VEEV) are endemic in North and South America, and infect humans and equine through mosquitoes. In addition, these viruses are highly infectious when aerosolized, making them potential biowarfare and bioterrorism agents. Currently, no approved vaccines or drugs are available for prevention and treatment. Extensive studies have been carried out to understand molecular mechanisms of virulence among New World alphaviruses. This review will focus on virus-encoded, interferon antagonizing proteins which play major role in determination of virulence of New World alphaviruses. Understanding of molecular mechanism of these proteins will shed light on development of broad-acting antivirals against New World alphaviruses.
Collapse
|
28
|
Complete coding sequences of eastern equine encephalitis virus and venezuelan equine encephalitis virus strains isolated from human cases. GENOME ANNOUNCEMENTS 2015; 3:3/2/e00243-15. [PMID: 25908124 PMCID: PMC4408325 DOI: 10.1128/genomea.00243-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We obtained the complete coding genome of an eastern equine encephalitis virus (EEEV) strain, EEEV V105-00210, and the complete genome of a Venezuelan equine encephalitis virus (VEEV) strain, VEEV INH-9813. They were obtained from human cases and are proposed as reference challenge strains for vaccine and therapeutic development in animal models.
Collapse
|
29
|
Ahola T, Karlin DG. Sequence analysis reveals a conserved extension in the capping enzyme of the alphavirus supergroup, and a homologous domain in nodaviruses. Biol Direct 2015; 10:16. [PMID: 25886938 PMCID: PMC4392871 DOI: 10.1186/s13062-015-0050-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 03/24/2015] [Indexed: 12/16/2022] Open
Abstract
Background Members of the alphavirus supergroup include human pathogens such as chikungunya virus, hepatitis E virus and rubella virus. They encode a capping enzyme with methyltransferase-guanylyltransferase (MTase-GTase) activity, which is an attractive drug target owing to its unique mechanism. However, its experimental study has proven very difficult. Results We examined over 50 genera of viruses by sequence analyses. Earlier studies showed that the MTase-GTase contains a “Core” region conserved in sequence. We show that it is followed by a long extension, which we termed “Iceberg” region, whose secondary structure, but not sequence, is strikingly conserved throughout the alphavirus supergroup. Sequence analyses strongly suggest that the minimal capping domain corresponds to the Core and Iceberg regions combined, which is supported by earlier experimental data. The Iceberg region contains all known membrane association sites that contribute to the assembly of viral replication factories. We predict that it may also contain an overlooked, widely conserved membrane-binding amphipathic helix. Unexpectedly, we detected a sequence homolog of the alphavirus MTase-GTase in taxa related to nodaviruses and to chronic bee paralysis virus. The presence of a capping enzyme in nodaviruses is biologically consistent, since they have capped genomes but replicate in the cytoplasm, where no cellular capping enzyme is present. The putative MTase-GTase domain of nodaviruses also contains membrane-binding sites that may drive the assembly of viral replication factories, revealing an unsuspected parallel with the alphavirus supergroup. Conclusions Our work will guide the functional analysis of the alphaviral MTase-GTase and the production of domains for structure determination. The identification of a homologous domain in a simple model system, nodaviruses, which replicate in numerous eukaryotic cell systems (yeast, flies, worms, mammals, and plants), can further help crack the function and structure of the enzyme. Reviewers This article was reviewed by Valerian Dolja, Eugene Koonin and Sebastian Maurer-Stroh. Electronic supplementary material The online version of this article (doi:10.1186/s13062-015-0050-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tero Ahola
- Department of Food and Environmental Sciences, University of Helsinki, 00014, Helsinki, Finland.
| | - David G Karlin
- Department of Zoology, University of Oxford, Oxford, OX1 3PS, UK. .,The Division of Structural Biology, Henry Wellcome Building, Roosevelt Drive, Oxford, OX3 7BN, UK.
| |
Collapse
|
30
|
Novel indole-2-carboxamide compounds are potent broad-spectrum antivirals active against western equine encephalitis virus in vivo. J Virol 2014; 88:11199-214. [PMID: 25031353 DOI: 10.1128/jvi.01671-14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Neurotropic alphaviruses, including western, eastern, and Venezuelan equine encephalitis viruses, cause serious and potentially fatal central nervous system infections in humans for which no currently approved therapies exist. We previously identified a series of thieno[3,2-b]pyrrole derivatives as novel inhibitors of neurotropic alphavirus replication, using a cell-based phenotypic assay (W. Peng et al., J. Infect. Dis. 199:950-957, 2009, doi:http://dx.doi.org/10.1086/597275), and subsequently developed second- and third-generation indole-2-carboxamide derivatives with improved potency, solubility, and metabolic stability (J. A. Sindac et al., J. Med. Chem. 55:3535-3545, 2012, doi:http://dx.doi.org/10.1021/jm300214e; J. A. Sindac et al., J. Med. Chem. 56:9222-9241, 2013, http://dx.doi.org/10.1021/jm401330r). In this report, we describe the antiviral activity of the most promising third-generation lead compound, CCG205432, and closely related analogs CCG206381 and CCG209023. These compounds have half-maximal inhibitory concentrations of ∼1 μM and selectivity indices of >100 in cell-based assays using western equine encephalitis virus replicons. Furthermore, CCG205432 retains similar potency against fully infectious virus in cultured human neuronal cells. These compounds show broad inhibitory activity against a range of RNA viruses in culture, including members of the Togaviridae, Bunyaviridae, Picornaviridae, and Paramyxoviridae families. Although their exact molecular target remains unknown, mechanism-of-action studies reveal that these novel indole-based compounds target a host factor that modulates cap-dependent translation. Finally, we demonstrate that both CCG205432 and CCG209023 dampen clinical disease severity and enhance survival of mice given a lethal western equine encephalitis virus challenge. These studies demonstrate that indole-2-carboxamide compounds are viable candidates for continued preclinical development as inhibitors of neurotropic alphaviruses and, potentially, of other RNA viruses. IMPORTANCE There are currently no approved drugs to treat infections with alphaviruses. We previously identified a novel series of compounds with activity against these potentially devastating pathogens (J. A. Sindac et al., J. Med. Chem. 55:3535-3545, 2012, doi:http://dx.doi.org/10.1021/jm300214e; W. Peng et al., J. Infect. Dis. 199:950-957, 2009, doi:http://dx.doi.org/10.1086/597275; J. A. Sindac et al., J. Med. Chem. 56:9222-9241, 2013, http://dx.doi.org/10.1021/jm401330r). We have now produced third-generation compounds with enhanced potency, and this manuscript provides detailed information on the antiviral activity of these advanced-generation compounds, including activity in an animal model. The results of this study represent a notable achievement in the continued development of this novel class of antiviral inhibitors.
Collapse
|
31
|
Antiviral perspectives for chikungunya virus. BIOMED RESEARCH INTERNATIONAL 2014; 2014:631642. [PMID: 24955364 PMCID: PMC4052087 DOI: 10.1155/2014/631642] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 04/22/2014] [Accepted: 04/30/2014] [Indexed: 12/17/2022]
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne pathogen that has a major health impact in humans and causes acute febrile illness in humans accompanied by joint pains and, in many cases, persistent arthralgia lasting for weeks to years. CHIKV reemerged in 2005-2006 in several parts of the Indian Ocean islands and India after a gap of 32 years, causing millions of cases. The re-emergence of CHIKV has also resulted in numerous outbreaks in several countries in the eastern hemisphere, with a threat to further expand in the near future. However, there is no vaccine against CHIKV infection licensed for human use, and therapy for CHIKV infection is still mainly limited to supportive care as antiviral agents are yet in different stages of testing or development. In this review we explore the different perspectives for chikungunya treatment and the effectiveness of these treatment regimens and discuss the scope for future directions.
Collapse
|
32
|
Hülseweh B, Rülker T, Pelat T, Langermann C, Frenzel A, Schirrmann T, Dübel S, Thullier P, Hust M. Human-like antibodies neutralizing Western equine encephalitis virus. MAbs 2014; 6:718-27. [PMID: 24518197 PMCID: PMC4011916 DOI: 10.4161/mabs.28170] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
This study describes the development of the first neutralizing antibodies against Western equine encephalitis virus (WEEV), a member of the genus Alphavirus. WEEV is transmitted by mosquitoes and can spread to the human central nervous system, causing symptoms ranging from mild febrile reactions to life-threatening encephalitis. WEEV has been classified as a biological warfare agent by the US Centers for Disease Control and Prevention. No anti-WEEV drugs are currently commercially available. Neutralizing antibodies are useful for the pre- and post-exposure treatment of WEEV infections. In this study, two immune antibody gene libraries were constructed from two macaques immunized with inactivated WEEV. Four antibodies were selected from these libraries and recloned as scFv-Fc, with a human Fc part. These antibodies bound WEEV specifically in ELISA with little or no cross-reaction with other alphaviruses. They were further analyzed by immunohistochemistry. All binders were suitable for the intracellular detection of WEEV particles. Neutralizing activity was determined in vitro. Three of the four antibodies were found to be neutralizing; about 1 ng/mL of the best antibody (ToR69–3A2) neutralized 50% of 5x104 TCID50/mL. Due to its human-like nature with a germinality index of 89% (VH) and 91% (VL), the ToR69–3A2 antibody is a promising candidate for future passive vaccine development.
Collapse
Affiliation(s)
- Birgit Hülseweh
- Wehrwissenschaftliches Institut für Schutztechnologien (WIS); ABC-Schutz; Munster, Germany
| | - Torsten Rülker
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie und Bioinformatik; Braunschweig, Germany
| | - Thibaut Pelat
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines; La Tronche, France
| | - Claudia Langermann
- Wehrwissenschaftliches Institut für Schutztechnologien (WIS); ABC-Schutz; Munster, Germany
| | - Andrè Frenzel
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie und Bioinformatik; Braunschweig, Germany
| | - Thomas Schirrmann
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie und Bioinformatik; Braunschweig, Germany
| | - Stefan Dübel
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie und Bioinformatik; Braunschweig, Germany
| | - Philippe Thullier
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines; La Tronche, France
| | - Michael Hust
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie und Bioinformatik; Braunschweig, Germany
| |
Collapse
|
33
|
Discovery of potent broad spectrum antivirals derived from marine actinobacteria. PLoS One 2013; 8:e82318. [PMID: 24349254 PMCID: PMC3857800 DOI: 10.1371/journal.pone.0082318] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 10/29/2013] [Indexed: 11/19/2022] Open
Abstract
Natural products provide a vast array of chemical structures to explore in the discovery of new medicines. Although secondary metabolites produced by microbes have been developed to treat a variety of diseases, including bacterial and fungal infections, to date there has been limited investigation of natural products with antiviral activity. In this report, we used a phenotypic cell-based replicon assay coupled with an iterative biochemical fractionation process to identify, purify, and characterize antiviral compounds produced by marine microbes. We isolated a compound from Streptomyces kaviengensis, a novel actinomycetes isolated from marine sediments obtained off the coast of New Ireland, Papua New Guinea, which we identified as antimycin A1a. This compound displays potent activity against western equine encephalitis virus in cultured cells with half-maximal inhibitory concentrations of less than 4 nM and a selectivity index of greater than 550. Our efforts also revealed that several antimycin A analogues display antiviral activity, and mechanism of action studies confirmed that these Streptomyces-derived secondary metabolites function by inhibiting the cellular mitochondrial electron transport chain, thereby suppressing de novo pyrimidine synthesis. Furthermore, we found that antimycin A functions as a broad spectrum agent with activity against a wide range of RNA viruses in cultured cells, including members of the Togaviridae, Flaviviridae, Bunyaviridae, Picornaviridae, and Paramyxoviridae families. Finally, we demonstrate that antimycin A reduces central nervous system viral titers, improves clinical disease severity, and enhances survival in mice given a lethal challenge with western equine encephalitis virus. Our results provide conclusive validation for using natural product resources derived from marine microbes as source material for antiviral drug discovery, and they indicate that host mitochondrial electron transport is a viable target for the continued development of broadly active antiviral compounds.
Collapse
|
34
|
Lundberg L, Pinkham C, Baer A, Amaya M, Narayanan A, Wagstaff KM, Jans DA, Kehn-Hall K. Nuclear import and export inhibitors alter capsid protein distribution in mammalian cells and reduce Venezuelan Equine Encephalitis Virus replication. Antiviral Res 2013; 100:662-72. [PMID: 24161512 DOI: 10.1016/j.antiviral.2013.10.004] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 10/11/2013] [Accepted: 10/15/2013] [Indexed: 11/22/2022]
Abstract
Targeting host responses to invading viruses has been the focus of recent antiviral research. Venezuelan Equine Encephalitis Virus (VEEV) is able to modulate host transcription and block nuclear trafficking at least partially due to its capsid protein forming a complex with the host proteins importin α/β1 and CRM1. We hypothesized that disrupting the interaction of capsid with importin α/β1 or the interaction of capsid with CRM1 would alter capsid localization, thereby lowering viral titers in vitro. siRNA mediated knockdown of importin α, importin β1, and CRM1 altered capsid localization, confirming their role in modulating capsid trafficking. Mifepristone and ivermectin, inhibitors of importin α/β-mediated import, were able to reduce nuclear-associated capsid, while leptomycin B, a potent CRM1 inhibitor, confined capsid to the nucleus. In addition to altering the level and distribution of capsid, the three inhibitors were able to reduce viral titers in a relevant mammalian cell line with varying degrees of efficacy. The inhibitors were also able to reduce the cytopathic effects associated with VEEV infection, hinting that nuclear import inhibitors may be protecting cells from apoptosis in addition to disrupting the function of an essential viral protein. Our results confirm that VEEV uses host importins and exportins during part of its life cycle. Further, it suggests that temporarily targeting host proteins that are hijacked for use by viruses is a viable antiviral therapy.
Collapse
Affiliation(s)
- Lindsay Lundberg
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, VA, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Spurgers KB, Hurt CR, Cohen JW, Eccelston LT, Lind CM, Lingappa VR, Glass PJ. Validation of a cell-based ELISA as a screening tool identifying anti-alphavirus small-molecule inhibitors. J Virol Methods 2013; 193:226-31. [DOI: 10.1016/j.jviromet.2013.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 05/17/2013] [Accepted: 06/03/2013] [Indexed: 12/29/2022]
|
36
|
Crystal structure of aura virus capsid protease and its complex with dioxane: new insights into capsid-glycoprotein molecular contacts. PLoS One 2012; 7:e51288. [PMID: 23251484 PMCID: PMC3522669 DOI: 10.1371/journal.pone.0051288] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Accepted: 11/01/2012] [Indexed: 12/25/2022] Open
Abstract
The nucleocapsid core interaction with endodomains of glycoproteins plays a critical role in the alphavirus life cycle that is essential to virus budding. Recent cryo-electron microscopy (cryo-EM) studies provide structural insights into key interactions between capsid protein (CP) and trans-membrane glycoproteins E1 and E2. CP possesses a chymotrypsin-like fold with a hydrophobic pocket at the surface responsible for interaction with glycoproteins. In the present study, crystal structures of the protease domain of CP from Aura virus and its complex with dioxane were determined at 1.81 and 1.98 Å resolution respectively. Due to the absence of crystal structures, homology models of E1 and E2 from Aura virus were generated. The crystal structure of CP and structural models of E1 and E2 were fitted into the cryo-EM density map of Venezuelan equine encephalitis virus (VEEV) for detailed analysis of CP-glycoprotein interactions. Structural analysis revealed that the E2 endodomain consists of a helix-loop-helix motif where the loop region fits into the hydrophobic pocket of CP. Our studies suggest that Cys397, Cys418 and Tyr401 residues of E2 are involved in stabilizing the structure of E2 endodomain. Density map fitting analysis revealed that Pro405, a conserved E2 residue is present in the loop region of the E2 endodomain helix-loop-helix structure and makes intermolecular hydrophobic contacts with the capsid. In the Aura virus capsid protease (AVCP)-dioxane complex structure, dioxane occupies the hydrophobic pocket on CP and structurally mimics the hydrophobic pyrollidine ring of Pro405 in the loop region of E2.
Collapse
|
37
|
Erwin-Cohen R, Porter A, Pittman P, Rossi C, Dasilva L. Host responses to live-attenuated Venezuelan equine encephalitis virus (TC-83): comparison of naïve, vaccine responder and nonresponder to TC-83 challenge in human peripheral blood mononuclear cells. Hum Vaccin Immunother 2012; 8:1053-65. [PMID: 22617845 DOI: 10.4161/hv.20300] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Venezuelan equine encephalitis virus (VEEV) is a positive-strand RNA Alphavirus endemic in Central and South America, and the causative agent of fatal encephalitis in humans. In an effort to better understand the mechanisms of infection, including differences between people who produce a neutralizing antibody response to the vaccine and those who do not, we performed whole genome transcriptional analysis in human PBMCs exposed in vitro to the live-attenuated vaccine strain of VEEV, TC-83. We compared the molecular responses in cells from three groups of individuals: naïve; previously vaccinated individuals who developed a neutralizing antibody response to the vaccine (responders); and those who did not develop a neutralizing antibody response to the vaccine (nonresponders). Overall, the changes in gene expression were more intense for the naïve group after TC-83 challenge and least potent in the nonresponder group. The main canonical pathways revealed the involvement of interferon and interferon-induced pathways, as well as toll-like receptors TLR- and interleukin (IL)-12-related pathways. HLA class II genotype and suppression of transcript expression for TLR2, TLR4 and TLR8 in the nonresponder group may help explain the lack of vaccine response in this study group. Because TL3 and TLR7 transcripts were elevated in all study groups, these factors may be indicators of the infection and not the immunological state of the individuals. Biomarkers were identified that differentiate between the vaccine responder and the vaccine nonresponder groups. The identified biomarkers were contrasted against transcripts that were unique to the naïve population alone upon induction with TC-83. Biomarker analysis allowed for the discernment between the naïve (innate) responses; the responder (recall) responses; and the nonresponder (alternative) changes to gene transcription that were caused by infection with TC-83. The study also points to the existence of HLA haplotypes that may discriminate between vaccine low- and high-responder phenotypes.
Collapse
Affiliation(s)
- Rebecca Erwin-Cohen
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Center for Aerobiological Sciences, 1425 Porter Street, Room 821, Frederick, MD 21702, USA.
| | | | | | | | | |
Collapse
|
38
|
Bowen DM, Lewis JA, Lu W, Schein CH. Simplifying complex sequence information: a PCP-consensus protein binds antibodies against all four Dengue serotypes. Vaccine 2012; 30:6081-7. [PMID: 22863657 DOI: 10.1016/j.vaccine.2012.07.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Revised: 07/13/2012] [Accepted: 07/18/2012] [Indexed: 12/15/2022]
Abstract
Designing proteins that reflect the natural variability of a pathogen is essential for developing novel vaccines and drugs. Flaviviruses, including Dengue (DENV) and West Nile (WNV), evolve rapidly and can "escape" neutralizing monoclonal antibodies by mutation. Designing antigens that represent many distinct strains is important for DENV, where infection with a strain from one of the four serotypes may lead to severe hemorrhagic disease on subsequent infection with a strain from another serotype. Here, a DENV physicochemical property (PCP)-consensus sequence was derived from 671 unique sequences from the Flavitrack database. PCP-consensus proteins for domain 3 of the envelope protein (EdomIII) were expressed from synthetic genes in Escherichia coli. The ability of the purified consensus proteins to bind polyclonal antibodies generated in response to infection with strains from each of the four DENV serotypes was determined. The initial consensus protein bound antibodies from DENV-1-3 in ELISA and Western blot assays. This sequence was altered in 3 steps to incorporate regions of maximum variability, identified as significant changes in the PCPs, characteristic of DENV-4 strains. The final protein was recognized by antibodies against all four serotypes. Two amino acids essential for efficient binding to all DENV antibodies are part of a discontinuous epitope previously defined for a neutralizing monoclonal antibody. The PCP-consensus method can significantly reduce the number of experiments required to define a multivalent antigen, which is particularly important when dealing with pathogens that must be tested at higher biosafety levels.
Collapse
Affiliation(s)
- David M Bowen
- Computational Biology, Sealy Center for Structural Biology and Molecular Biophysics, Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555-0857, United States
| | | | | | | |
Collapse
|
39
|
Kehn-Hall K, Narayanan A, Lundberg L, Sampey G, Pinkham C, Guendel I, Van Duyne R, Senina S, Schultz KL, Stavale E, Aman MJ, Bailey C, Kashanchi F. Modulation of GSK-3β activity in Venezuelan equine encephalitis virus infection. PLoS One 2012; 7:e34761. [PMID: 22496857 PMCID: PMC3319612 DOI: 10.1371/journal.pone.0034761] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 03/08/2012] [Indexed: 11/18/2022] Open
Abstract
Alphaviruses, including Venezuelan Equine Encephalitis Virus (VEEV), cause disease in both equine and humans that exhibit overt encephalitis in a significant percentage of cases. Features of the host immune response and tissue-specific responses may contribute to fatal outcomes as well as the development of encephalitis. It has previously been shown that VEEV infection of mice induces transcription of pro-inflammatory cytokines genes (e.g., IFN-γ, IL-6, IL-12, iNOS and TNF-α) within 6 h. GSK-3β is a host protein that is known to modulate pro-inflammatory gene expression and has been a therapeutic target in neurodegenerative disorders such as Alzheimer's. Hence inhibition of GSK-3β in the context of encephalitic viral infections has been useful in a neuroprotective capacity. Small molecule GSK-3β inhibitors and GSK-3β siRNA experiments indicated that GSK-3β was important for VEEV replication. Thirty-eight second generation BIO derivatives were tested and BIOder was found to be the most potent inhibitor, with an IC50 of ∼0.5 µM and a CC50 of >100 µM. BIOder was a more potent inhibitor of GSK-3β than BIO, as demonstrated through in vitro kinase assays from uninfected and infected cells. Size exclusion chromatography experiments demonstrated that GSK-3β is found in three distinct complexes in VEEV infected cells, whereas GSK-3β is only present in one complex in uninfected cells. Cells treated with BIOder demonstrated an increase in the anti-apoptotic gene, survivin, and a decrease in the pro-apoptotic gene, BID, suggesting that modulation of pro- and anti-apoptotic genes contributes to the protective effect of BIOder treatment. Finally, BIOder partially protected mice from VEEV induced mortality. Our studies demonstrate the utility of GSK-3β inhibitors for modulating VEEV infection.
Collapse
Affiliation(s)
- Kylene Kehn-Hall
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia, United States of America
| | - Aarthi Narayanan
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia, United States of America
| | - Lindsay Lundberg
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia, United States of America
| | - Gavin Sampey
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia, United States of America
| | - Chelsea Pinkham
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia, United States of America
| | - Irene Guendel
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia, United States of America
| | - Rachel Van Duyne
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia, United States of America
- Department of Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, D.C., United States of America
| | - Svetlana Senina
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia, United States of America
| | - Kimberly L. Schultz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Eric Stavale
- Integrated Biotherapeutics Inc., Gaithersburg, Maryland, United States of America
| | - M. Javad Aman
- Integrated Biotherapeutics Inc., Gaithersburg, Maryland, United States of America
| | - Charles Bailey
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia, United States of America
| | - Fatah Kashanchi
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia, United States of America
- * E-mail:
| |
Collapse
|
40
|
Sampath A, Metz M, Stundick M, Larsen JC. State-of-the-art therapeutic medical countermeasures for viral threat agents. Biosecur Bioterror 2011; 9:351-60. [PMID: 22053938 DOI: 10.1089/bsp.2011.0047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
In recent years, there has been an increase in the perceived threat of biological agents being used against civilian populations. This has prompted an urgent need for the development and procurement of medical countermeasures (MCMs) against highly pathogenic viruses that can prevent morbidity and mortality from infections caused by these agents. To date, antiviral drug development has been largely focused on clinically prevalent chronic infections due to their commercial viability. This has left a huge gap in the drug development path for acute infections of biodefense importance. In this review, we discuss the antiviral research and development initiatives focusing specifically on poxviruses, filoviruses, and equine encephalitis viruses (EEV). We discuss the benefits and technical challenges in the current development strategies and the hurdles in the licensure path for MCMs against these highly pathogenic viruses under the FDA Animal Rule, and we provide recommendations for the path forward.
Collapse
Affiliation(s)
- Aruna Sampath
- Science Applications International Corporation, Frederick, Maryland, USA
| | | | | | | |
Collapse
|
41
|
Aggarwal M, Dhindwal S, Pratap S, Kuhn RJ, Kumar P, Tomar S. Crystallization, high-resolution data collection and preliminary crystallographic analysis of Aura virus capsid protease and its complex with dioxane. Acta Crystallogr Sect F Struct Biol Cryst Commun 2011; 67:1394-8. [PMID: 22102240 DOI: 10.1107/s174430911103404x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 08/19/2011] [Indexed: 08/30/2023]
Abstract
The C-terminal protease domain of capsid protein from Aura virus expressed in a bacterial expression system has been purified to homogeneity and crystallized. Crystals suitable for X-ray diffraction analysis were obtained by the vapour-diffusion method using 0.1 M bis-tris and polyethylene glycol monomethyl ether 2000. Crystals of the C-terminal protease domain of capsid protein in complex with dioxane were also produced and crystal data were obtained. Both crystals belonged to space group C2, with unit-cell parameters a = 79.6, b = 35.2, c = 49.5 Å. High-resolution data sets were collected to a resolution of 1.81 Å for the native protein and 1.98 Å for the complex. Preliminary crystallographic studies suggested the presence of a single molecule in the crystallographic asymmetric unit, with a solvent content of 38.5%.
Collapse
Affiliation(s)
- Megha Aggarwal
- Department of Biotechnology, Indian Institute of Technology, Roorkee, Roorkee 247 667, India
| | | | | | | | | | | |
Collapse
|
42
|
Goodchild SA, O'Brien LM, Steven J, Muller MR, Lanning OJ, Logue CH, D'Elia RV, Phillpotts RJ, Perkins SD. A humanised murine monoclonal antibody with broad serogroup specificity protects mice from challenge with Venezuelan equine encephalitis virus. Antiviral Res 2011; 90:1-8. [PMID: 21310183 DOI: 10.1016/j.antiviral.2011.01.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 01/31/2011] [Accepted: 01/31/2011] [Indexed: 12/21/2022]
Abstract
In murine models of Venezuelan equine encephalitis virus (VEEV) infection, the neutralising monoclonal antibody 1A3B-7 has been shown to be effective in passive protection from challenge by the aerosol route with serogroups I, II and Mucambo virus (formally VEE complex subtype IIIA). This antibody is able to bind to all serogroups of the VEEV complex when used in ELISA and therefore is an excellent candidate for protein engineering in order to derive a humanised molecule suitable for therapeutic use in humans. A Complementarity Determining Region (CDR) grafting approach using human germline IgG frameworks was used to produce a panel of humanised variants of 1A3B-7, from which a single candidate molecule with retained binding specificity was identified. Evaluation of humanised 1A3B-7 (Hu1A3B-7) in in vitro studies indicated that Hu1A3B-7 retained both broad specificity and neutralising activity. Furthermore, in vivo experiments showed that Hu1A3B-7 successfully protected mice against lethal subcutaneous and aerosol challenges with VEEV strain TrD (serogroup I). Hu1A3B-7 is therefore a promising candidate for the future development of a broad-spectrum antiviral therapy to treat VEEV disease in humans.
Collapse
Affiliation(s)
- Sarah A Goodchild
- Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire SP4 0JQ, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Bhomia M, Balakathiresan N, Sharma A, Gupta P, Biswas R, Maheshwari R. Analysis of microRNAs induced by Venezuelan equine encephalitis virus infection in mouse brain. Biochem Biophys Res Commun 2010; 395:11-6. [PMID: 20303929 DOI: 10.1016/j.bbrc.2010.03.091] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Accepted: 03/16/2010] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNA) are small RNA (approximately 22nts) molecules that are expressed endogenously in cells and play an important role in regulating gene expression. Recent studies have shown that cellular miRNA plays a very important role in the pathogenesis of viral infection. Venezuelan equine encephalitis virus (VEEV) is an RNA virus and is a member of the genus Alphavirus in the family Togaviridae. VEEV is infectious in aerosol form and is a potential biothreat agent. In this study, we report for the first time that VEEV infection in mice brain causes modulation of miRNA expression. Pathway analyses showed that majority of these miRNAs are involved in the neuronal development and function. Target gene prediction of the modulated miRNAs correlates with our recently reported mRNA expression in VEEV infected mice brain.
Collapse
Affiliation(s)
- Manish Bhomia
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | | | | | | | | | | |
Collapse
|