101
|
Sergeev AA, Kabanov AS, Bulychev LE, Sergeev AA, Pyankov OV, Bodnev SA, Galahova DO, Zamedyanskaya AS, Titova KA, Glotova TI, Taranov OS, Omigov VV, Shishkina LN, Agafonov AP, Sergeev AN. Using the Ground Squirrel (Marmota bobak) as an Animal Model to Assess Monkeypox Drug Efficacy. Transbound Emerg Dis 2017; 64:226-236. [PMID: 25944444 DOI: 10.1111/tbed.12364] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Indexed: 11/29/2022]
Abstract
In experiments to study the sensitivity of ground squirrels (Marmota bobak) to monkeypox virus (MPXV) at intranasal challenge, expressed pox-like clinical symptoms (hyperthermia, lymphadenitis, skin rash all over the body and mucous membranes and others) were observed 7-9 days post-infection. The 50% infective dose (ID50 ) of MPXV for these marmots determined by the presence of clinical signs of the disease was 2.2 log10 PFU. Some diseased marmots (about 40%) died 13-22 days post-infection, and the mortality rate was weakly dependent on MPXV infective dose. Lungs with trachea were primary target organs of marmots challenged intranasally (with ~30 ID50 ). The pathogen got to secondary target organs of the animals mainly via the lymphatic way (with replication in bifurcation lymph nodes). Lungs with trachea, nasal mucosa and skin were the organs where the maximum MPXV amounts accumulated in these animals. Evidences of the pathogen presence and replication were revealed in these and subcutaneously infected marmots in the traditional primary target cells for MPXV (macrophages and respiratory tract epitheliocytes), as well as in some other cells (endotheliocytes, plasmocytes, fibroblasts, reticular and smooth muscle cells). Our use of this animal species to assess the antiviral efficacy of some drugs demonstrated the agreement of the obtained results with those described in scientific literature, which opens up the prospects of using marmots as animal models for monkeypox to develop therapeutic and preventive anti-smallpox drugs.
Collapse
Affiliation(s)
- A A Sergeev
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology VECTOR, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - A S Kabanov
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology VECTOR, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - L E Bulychev
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology VECTOR, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - A A Sergeev
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology VECTOR, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - O V Pyankov
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology VECTOR, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - S A Bodnev
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology VECTOR, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - D O Galahova
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology VECTOR, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - A S Zamedyanskaya
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology VECTOR, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - K A Titova
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology VECTOR, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - T I Glotova
- State Scientific Establishment - Institute of Experimental Veterinary Science of Siberia and the Far East Russian Academy of Agricultural Sciences, Krasnoobsk, Russia
| | - O S Taranov
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology VECTOR, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - V V Omigov
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology VECTOR, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - L N Shishkina
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology VECTOR, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - A P Agafonov
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology VECTOR, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - A N Sergeev
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology VECTOR, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| |
Collapse
|
102
|
Sergeev AA, Kabanov AS, Bulychev LE, Sergeev AA, Pyankov OV, Bodnev SA, Galahova DO, Zamedyanskaya AS, Titova KA, Glotov AG, Taranov OS, Omigov VV, Shishkina LN, Agafonov AP, Sergeev AN. The Possibility of Using the ICR Mouse as an Animal Model to Assess Antimonkeypox Drug Efficacy. Transbound Emerg Dis 2016; 63:e419-30. [PMID: 25597343 DOI: 10.1111/tbed.12323] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Indexed: 12/30/2022]
Abstract
As a result of the conducted experimental studies on intranasal challenge of ICR mice, rabbits and miniature pigs (even in the maximum variant) with the doses of 4.0-5.5 lg PFU of monkeypox virus (MPXV), some clinical signs such as purulent conjunctivitis, blepharitis and ruffled fur were found only in mice. The 50% infective dose (C ID50 ) of MPXV for these animals estimated by the presence of external clinical signs was 4.8 lg PFU, and L ID50 estimated by the virus presence in the lungs of mice 7 days post-infection taking into account its 10% application in the animal respiratory tract was 1.4 lg PFU. When studying the dynamics of MPXV propagation in mice challenged intranasally with 25 L ID50 of MPXV, the maximum pathogen accumulation was revealed in nasal cavity, lungs and brain: 5.7 ± 0.1, 5.5 ± 0.1 and 5.3 ± 0.3 lg PFU/ml, respectively. The pathomorphological examination of these animals revealed the presence and replication of the pathogen in the traditional primary target cells for MPXV (mononuclear phagocyte system cells and respiratory tract epitheliocytes) as well as in some other types of cells (endothelial cells, reticular cells, connective tissue cells). Our use of these animals to assess the antiviral efficacy of some drugs demonstrated the agreement of the results (a significant positive effect of NIOCH-14 and ST-246) with those described in scientific literature, which opens up the prospects of using ICR mice as animal models for monkeypox to develop preventive antismallpox drugs.
Collapse
Affiliation(s)
- Al A Sergeev
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - A S Kabanov
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - L E Bulychev
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - Ar A Sergeev
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - O V Pyankov
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - S A Bodnev
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - D O Galahova
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - A S Zamedyanskaya
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - K A Titova
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - A G Glotov
- State Scientific Establishment - Institute of Experimental Veterinary Science of Siberia and the Far East Russian Academy of Agricultural Sciences, Krasnoobsk, Russia
| | - O S Taranov
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - V V Omigov
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - L N Shishkina
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - A P Agafonov
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| | - A N Sergeev
- Federal Budgetary Research Institution - State Research Center of Virology and Biotechnology Vector, Federal Service for Surveillance on Consumer Rights Protection and Human Well-being, Koltsovo, Russia
| |
Collapse
|
103
|
Johnson RF, Hammoud DA, Perry DL, Solomon J, Moore IN, Lackemeyer MG, Bohannon JK, Sayre PJ, Minai M, Papaneri AB, Hagen KR, Janosko KB, Jett C, Cooper K, Blaney JE, Jahrling PB. Exposure of rhesus monkeys to cowpox virus Brighton Red by large-particle aerosol droplets results in an upper respiratory tract disease. J Gen Virol 2016; 97:1942-1954. [PMID: 27166137 PMCID: PMC5764124 DOI: 10.1099/jgv.0.000501] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 05/07/2016] [Indexed: 01/13/2023] Open
Abstract
We previously demonstrated that small-particle (0.5-3.0 µm) aerosol infection of rhesus monkeys (Macaca mulatta) with cowpox virus (CPXV)-Brighton Red (BR) results in fulminant respiratory tract disease characterized by severe lung parenchymal pathology but only limited systemic virus dissemination and limited classic epidermal pox-like lesion development (Johnson et al., 2015). Based on these results, and to further develop CPXV as an improved model of human smallpox, we evaluated a novel large-particle aerosol (7.0-9.0 µm) exposure of rhesus monkeys to CPXV-BR and monitored for respiratory tract disease by serial computed tomography (CT). As expected, the upper respiratory tract and large airways were the major sites of virus-induced pathology following large-particle aerosol exposure. Large-particle aerosol CPXV exposure of rhesus macaques resulted in severe upper airway and large airway pathology with limited systemic dissemination.
Collapse
Affiliation(s)
- Reed F. Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Dima A. Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Donna L. Perry
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Jeffrey Solomon
- Clinical Research Directorate/Clinical Monitoring Research Program Leidos Biomedical Research, Inc, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Ian N. Moore
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew G. Lackemeyer
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Jordan K. Bohannon
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Philip J. Sayre
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Amy B. Papaneri
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Katie R. Hagen
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Krisztina B. Janosko
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Catherine Jett
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Kurt Cooper
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Joseph E. Blaney
- Office of the Scientific Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter B. Jahrling
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| |
Collapse
|
104
|
Falendysz EA, Lopera JG, Lorenzsonn F, Salzer JS, Hutson CL, Doty J, Gallardo-Romero N, Carroll DS, Osorio JE, Rocke TE. Further Assessment of Monkeypox Virus Infection in Gambian Pouched Rats (Cricetomys gambianus) Using In Vivo Bioluminescent Imaging. PLoS Negl Trop Dis 2015; 9:e0004130. [PMID: 26517839 PMCID: PMC4627722 DOI: 10.1371/journal.pntd.0004130] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 09/09/2015] [Indexed: 02/06/2023] Open
Abstract
Monkeypox is a zoonosis clinically similar to smallpox in humans. Recent evidence has shown a potential risk of increased incidence in central Africa. Despite attempts to isolate the virus from wild rodents and other small mammals, no reservoir host has been identified. In 2003, Monkeypox virus (MPXV) was accidentally introduced into the U.S. via the pet trade and was associated with the Gambian pouched rat (Cricetomys gambianus). Therefore, we investigated the potential reservoir competence of the Gambian pouched rat for MPXV by utilizing a combination of in vivo and in vitro methods. We inoculated three animals by the intradermal route and three animals by the intranasal route, with one mock-infected control for each route. Bioluminescent imaging (BLI) was used to track replicating virus in infected animals and virological assays (e.g. real time PCR, cell culture) were used to determine viral load in blood, urine, ocular, nasal, oral, and rectal swabs. Intradermal inoculation resulted in clinical signs of monkeypox infection in two of three animals. One severely ill animal was euthanized and the other affected animal recovered. In contrast, intranasal inoculation resulted in subclinical infection in all three animals. All animals, regardless of apparent or inapparent infection, shed virus in oral and nasal secretions. Additionally, BLI identified viral replication in the skin without grossly visible lesions. These results suggest that Gambian pouched rats may play an important role in transmission of the virus to humans, as they are hunted for consumption and it is possible for MPXV-infected pouched rats to shed infectious virus without displaying overt clinical signs.
Collapse
Affiliation(s)
- Elizabeth A. Falendysz
- U.S. Geological Survey-National Wildlife Health Center, Madison, Wisconsin, United States of America
| | - Juan G. Lopera
- Department of Pathobiological Science, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Faye Lorenzsonn
- Department of Pathobiological Science, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Johanna S. Salzer
- Centers for Disease Control and Prevention, National Centers for Zoonotic and Vector-Borne and Enteric Diseases, Division of High Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, Atlanta, Georgia, United States of America
| | - Christina L. Hutson
- Centers for Disease Control and Prevention, National Centers for Zoonotic and Vector-Borne and Enteric Diseases, Division of High Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, Atlanta, Georgia, United States of America
| | - Jeffrey Doty
- Centers for Disease Control and Prevention, National Centers for Zoonotic and Vector-Borne and Enteric Diseases, Division of High Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, Atlanta, Georgia, United States of America
| | - Nadia Gallardo-Romero
- Centers for Disease Control and Prevention, National Centers for Zoonotic and Vector-Borne and Enteric Diseases, Division of High Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, Atlanta, Georgia, United States of America
| | - Darin S. Carroll
- Centers for Disease Control and Prevention, National Centers for Zoonotic and Vector-Borne and Enteric Diseases, Division of High Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, Atlanta, Georgia, United States of America
| | - Jorge E. Osorio
- Department of Pathobiological Science, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Tonie E. Rocke
- U.S. Geological Survey-National Wildlife Health Center, Madison, Wisconsin, United States of America
| |
Collapse
|
105
|
Mucker EM, Chapman J, Huzella LM, Huggins JW, Shamblin J, Robinson CG, Hensley LE. Susceptibility of Marmosets (Callithrix jacchus) to Monkeypox Virus: A Low Dose Prospective Model for Monkeypox and Smallpox Disease. PLoS One 2015; 10:e0131742. [PMID: 26147658 PMCID: PMC4492619 DOI: 10.1371/journal.pone.0131742] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 06/05/2015] [Indexed: 01/01/2023] Open
Abstract
Although current nonhuman primate models of monkeypox and smallpox diseases provide some insight into disease pathogenesis, they require a high titer inoculum, use an unnatural route of infection, and/or do not accurately represent the entire disease course. This is a concern when developing smallpox and/or monkeypox countermeasures or trying to understand host pathogen relationships. In our studies, we altered half of the test system by using a New World nonhuman primate host, the common marmoset. Based on dose finding studies, we found that marmosets are susceptible to monkeypox virus infection, produce a high viremia, and have pathological features consistent with smallpox and monkeypox in humans. The low dose (48 plaque forming units) required to elicit a uniformly lethal disease and the extended incubation (preclinical signs) are unique features among nonhuman primate models utilizing monkeypox virus. The uniform lethality, hemorrhagic rash, high viremia, decrease in platelets, pathology, and abbreviated acute phase are reflective of early-type hemorrhagic smallpox.
Collapse
Affiliation(s)
- Eric M. Mucker
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
- Tulane University School of Medicine, New Orleans, Louisianna, United States of America
| | - Jennifer Chapman
- Joint Pathology Center, Silver Spring, Maryland, United States of America
| | - Louis M. Huzella
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - John W. Huggins
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Joshua Shamblin
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Camenzind G. Robinson
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Lisa E. Hensley
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, Fort Detrick, Maryland, United States of America
| |
Collapse
|
106
|
Johnson RF, Hammoud DA, Lackemeyer MG, Yellayi S, Solomon J, Bohannon JK, Janosko KB, Jett C, Cooper K, Blaney JE, Jahrling PB. Small particle aerosol inoculation of cowpox Brighton Red in rhesus monkeys results in a severe respiratory disease. Virology 2015; 481:124-35. [PMID: 25776759 PMCID: PMC4535421 DOI: 10.1016/j.virol.2015.02.044] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/17/2015] [Accepted: 02/18/2015] [Indexed: 10/23/2022]
Abstract
Cowpox virus (CPXV) inoculation of nonhuman primates (NHPs) has been suggested as an alternate model for smallpox (Kramski et al., 2010, PLoS One, 5, e10412). Previously, we have demonstrated that intrabronchial inoculation of CPXV-Brighton Red (CPXV-BR) into cynomolgus monkeys resulted in a disease that shared many similarities to smallpox; however, severe respiratory tract disease was observed (Smith et al., 2011, J. Gen. Virol.). Here we describe the course of disease after small particle aerosol exposure of rhesus monkeys using computed tomography (CT) to monitor respiratory disease progression. Subjects developed a severe respiratory disease that was uniformly lethal at 5.7 log10 PFU of CPXV-BR. CT indicated changes in lung architecture that correlated with changes in peripheral blood monocytes and peripheral oxygen saturation. While the small particle aerosol inoculation route does not accurately mimic human smallpox, the data suggest that CT can be used as a tool to monitor real-time disease progression for evaluation of animal models for human diseases.
Collapse
Affiliation(s)
- Reed F Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Dima A Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew G Lackemeyer
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Srikanth Yellayi
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Jeffrey Solomon
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jordan K Bohannon
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Krisztina B Janosko
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Catherine Jett
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Kurt Cooper
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Joseph E Blaney
- Office of the Scientific Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter B Jahrling
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA; Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| |
Collapse
|
107
|
Franceschi V, Parker S, Jacca S, Crump RW, Doronin K, Hembrador E, Pompilio D, Tebaldi G, Estep RD, Wong SW, Buller MR, Donofrio G. BoHV-4-Based Vector Single Heterologous Antigen Delivery Protects STAT1(-/-) Mice from Monkeypoxvirus Lethal Challenge. PLoS Negl Trop Dis 2015; 9:e0003850. [PMID: 26086739 PMCID: PMC4473039 DOI: 10.1371/journal.pntd.0003850] [Citation(s) in RCA: 30] [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: 12/30/2014] [Accepted: 05/27/2015] [Indexed: 01/13/2023] Open
Abstract
Monkeypox virus (MPXV) is the etiological agent of human (MPX). It is an emerging orthopoxvirus zoonosis in the tropical rain forest of Africa and is endemic in the Congo-basin and sporadic in West Africa; it remains a tropical neglected disease of persons in impoverished rural areas. Interaction of the human population with wildlife increases human infection with MPX virus (MPXV), and infection from human to human is possible. Smallpox vaccination provides good cross-protection against MPX; however, the vaccination campaign ended in Africa in 1980, meaning that a large proportion of the population is currently unprotected against MPXV infection. Disease control hinges on deterring zoonotic exposure to the virus and, barring that, interrupting person-to-person spread. However, there are no FDA-approved therapies against MPX, and current vaccines are limited due to safety concerns. For this reason, new studies on pathogenesis, prophylaxis and therapeutics are still of great interest, not only for the scientific community but also for the governments concerned that MPXV could be used as a bioterror agent. In the present study, a new vaccination strategy approach based on three recombinant bovine herpesvirus 4 (BoHV-4) vectors, each expressing different MPXV glycoproteins, A29L, M1R and B6R were investigated in terms of protection from a lethal MPXV challenge in STAT1 knockout mice. BoHV-4-A-CMV-A29LgD106ΔTK, BoHV-4-A-EF1α-M1RgD106ΔTK and BoHV-4-A-EF1α-B6RgD106ΔTK were successfully constructed by recombineering, and their capacity to express their transgene was demonstrated. A small challenge study was performed, and all three recombinant BoHV-4 appeared safe (no weight-loss or obvious adverse events) following intraperitoneal administration. Further, BoHV-4-A-EF1α-M1RgD106ΔTK alone or in combination with BoHV-4-A-CMV-A29LgD106ΔTK and BoHV-4-A-EF1α-B6RgD106ΔTK, was shown to be able to protect, 100% alone and 80% in combination, STAT1(-/-) mice against mortality and morbidity. This work demonstrated the efficacy of BoHV-4 based vectors and the use of BoHV-4 as a vaccine-vector platform.
Collapse
Affiliation(s)
| | - Scott Parker
- Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Sarah Jacca
- Department of Medical-Veterinary Science, University of Parma, Parma, Italy
| | - Ryan W. Crump
- Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Konstantin Doronin
- Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Edguardo Hembrador
- Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Daniela Pompilio
- Department of Medical-Veterinary Science, University of Parma, Parma, Italy
| | - Giulia Tebaldi
- Department of Medical-Veterinary Science, University of Parma, Parma, Italy
| | - Ryan D. Estep
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Scott W. Wong
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Mark R. Buller
- Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Gaetano Donofrio
- Department of Medical-Veterinary Science, University of Parma, Parma, Italy
| |
Collapse
|
108
|
Rehfeld IS, Guedes MIMC, Fraiha ALS, Costa AG, Matos ACD, Fiúza ATL, Lobato ZIP. Vaccinia virus Transmission through Experimentally Contaminated Milk Using a Murine Model. PLoS One 2015; 10:e0127350. [PMID: 26000966 PMCID: PMC4441451 DOI: 10.1371/journal.pone.0127350] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/14/2015] [Indexed: 11/19/2022] Open
Abstract
Bovine vaccinia (BV) is a zoonosis caused by Vaccinia virus (VACV), which affects dairy cattle and humans. Previous studies have detected the presence of viable virus particles in bovine milk samples naturally and experimentally contaminated with VACV. However, it is not known whether milk contaminated with VACV could be a route of viral transmission. However, anti-Orthopoxvirus antibodies were detected in humans from BV endemic areas, whom had no contact with affected cows, which suggest that other VACV transmission routes are possible, such as consumption of contaminated milk and dairy products. Therefore, it is important to study the possibility of VACV transmission by contaminated milk. This study aimed to examine VACV transmission, pathogenesis and shedding in mice orally inoculated with experimentally contaminated milk. Thirty mice were orally inoculated with milk containing 107 PFU/ml of VACV, and ten mice were orally inoculated with uncontaminated milk. Clinical examinations were performed for 30 consecutive days, and fecal samples and oral swabs (OSs) were collected every other day. Mice were euthanized on predetermined days, and tissue and blood samples were collected. Nested-PCR, plaque reduction neutralization test (PRNT), viral isolation, histopathology, and immunohistochemistry (IHC) methods were performed on the collected samples. No clinical changes were observed in the animals. Viral DNA was detected in feces, blood, OSs and tissues, at least in one of the times tested. The lungs displayed moderate to severe interstitial lymphohistiocytic infiltrates, and only the heart, tonsils, tongue, and stomach did not show immunostaining at the IHC analysis. Neutralizing antibodies were detected at the 20th and 30th days post infection in 50% of infected mice. The results revealed that VACV contaminated milk could be a route of viral transmission in mice experimentally infected, showing systemic distribution and shedding through feces and oral mucosa, albeit without exhibiting any clinical signs.
Collapse
Affiliation(s)
- Izabelle Silva Rehfeld
- Laboratório de Pesquisa em Virologia Animal (LPVA), Departamento de Medicina Veterinária Preventiva, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maria Isabel Maldonado Coelho Guedes
- Laboratório de Pesquisa em Virologia Animal (LPVA), Departamento de Medicina Veterinária Preventiva, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Luiza Soares Fraiha
- Laboratório de Pesquisa em Virologia Animal (LPVA), Departamento de Medicina Veterinária Preventiva, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Aristóteles Gomes Costa
- Laboratório de Pesquisa em Virologia Animal (LPVA), Departamento de Medicina Veterinária Preventiva, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Carolina Diniz Matos
- Laboratório de Pesquisa em Virologia Animal (LPVA), Departamento de Medicina Veterinária Preventiva, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Aparecida Tatiane Lino Fiúza
- Laboratório de Patologia Veterinária, Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Zélia Inês Portela Lobato
- Laboratório de Pesquisa em Virologia Animal (LPVA), Departamento de Medicina Veterinária Preventiva, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
109
|
Tree JA, Hall G, Pearson G, Rayner E, Graham VA, Steeds K, Bewley KR, Hatch GJ, Dennis M, Taylor I, Roberts AD, Funnell SGP, Vipond J. Sequence of pathogenic events in cynomolgus macaques infected with aerosolized monkeypox virus. J Virol 2015; 89:4335-44. [PMID: 25653439 PMCID: PMC4442344 DOI: 10.1128/jvi.03029-14] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 01/26/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED To evaluate new vaccines when human efficacy studies are not possible, the FDA's "Animal Rule" requires well-characterized models of infection. Thus, in the present study, the early pathogenic events of monkeypox infection in nonhuman primates, a surrogate for variola virus infection, were characterized. Cynomolgus macaques were exposed to aerosolized monkeypox virus (10(5) PFU). Clinical observations, viral loads, immune responses, and pathological changes were examined on days 2, 4, 6, 8, 10, and 12 postchallenge. Viral DNA (vDNA) was detected in the lungs on day 2 postchallenge, and viral antigen was detected, by immunostaining, in the epithelium of bronchi, bronchioles, and alveolar walls. Lesions comprised rare foci of dysplastic and sloughed cells in respiratory bronchioles. By day 4, vDNA was detected in the throat, tonsil, and spleen, and monkeypox antigen was detected in the lung, hilar and submandibular lymph nodes, spleen, and colon. Lung lesions comprised focal epithelial necrosis and inflammation. Body temperature peaked on day 6, pox lesions appeared on the skin, and lesions, with positive immunostaining, were present in the lung, tonsil, spleen, lymph nodes, and colon. By day 8, vDNA was present in 9/13 tissues. Blood concentrations of interleukin 1ra (IL-1ra), IL-6, and gamma interferon (IFN-γ) increased markedly. By day 10, circulating IgG antibody concentrations increased, and on day 12, animals showed early signs of recovery. These results define early events occurring in an inhalational macaque monkeypox infection model, supporting its use as a surrogate model for human smallpox. IMPORTANCE Bioterrorism poses a major threat to public health, as the deliberate release of infectious agents, such smallpox or a related virus, monkeypox, would have catastrophic consequences. The development and testing of new medical countermeasures, e.g., vaccines, are thus priorities; however, tests for efficacy in humans cannot be performed because it would be unethical and field trials are not feasible. To overcome this, the FDA may grant marketing approval of a new product based upon the "Animal Rule," in which interventions are tested for efficacy in well-characterized animal models. Monkeypox virus infection of nonhuman primates (NHPs) presents a potential surrogate disease model for smallpox. Previously, the later stages of monkeypox infection were defined, but the early course of infection remains unstudied. Here, the early pathogenic events of inhalational monkeypox infection in NHPs were characterized, and the results support the use of this surrogate model for testing human smallpox interventions.
Collapse
Affiliation(s)
- J A Tree
- Microbiological Services, Public Health England, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - G Hall
- Microbiological Services, Public Health England, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - G Pearson
- Microbiological Services, Public Health England, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - E Rayner
- Microbiological Services, Public Health England, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - V A Graham
- Microbiological Services, Public Health England, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - K Steeds
- Microbiological Services, Public Health England, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - K R Bewley
- Microbiological Services, Public Health England, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - G J Hatch
- Microbiological Services, Public Health England, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - M Dennis
- Microbiological Services, Public Health England, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - I Taylor
- Microbiological Services, Public Health England, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - A D Roberts
- Microbiological Services, Public Health England, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - S G P Funnell
- Microbiological Services, Public Health England, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - J Vipond
- Microbiological Services, Public Health England, Porton Down, Salisbury, Wiltshire, United Kingdom
| |
Collapse
|
110
|
Earl PL, Americo JL, Cotter CA, Moss B. Comparative live bioluminescence imaging of monkeypox virus dissemination in a wild-derived inbred mouse (Mus musculus castaneus) and outbred African dormouse (Graphiurus kelleni). Virology 2015; 475:150-8. [PMID: 25462355 PMCID: PMC4280325 DOI: 10.1016/j.virol.2014.11.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 11/13/2014] [Accepted: 11/14/2014] [Indexed: 11/28/2022]
Abstract
Monkeypox virus belongs to the orthopoxvirus genus, infects rodents and monkeys in Africa, produces a smallpox-like zoonotic disease in humans, and has the potential for global spread and exploitation for bioterrorism. Several small animal models for studying monkeypox virus pathogenesis have been investigated. The African dormouse is a candidate natural host but is outbred and no immunological reagents exist. Although not a natural host, the CAST/EiJ mouse is inbred and animals and reagents are commercially available. We compared the dissemination of monkeypox virus by bioluminescence imaging in CAST/EiJ mice and dormice. In CAST/EiJ mice, intense replication occurred at the intranasal site of inoculation and virus spread rapidly to lungs and abdominal organs, which had a lower virus burden. Compared to CAST/EiJ mice, dormice exhibited a greater variation of virus spread, a slower time course, less replication in the head and chest, and more replication in abdominal organs prior to death.
Collapse
Affiliation(s)
- Patricia L Earl
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Jeffrey L Americo
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Catherine A Cotter
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
111
|
Postchallenge administration of brincidofovir protects healthy and immune-deficient mice reconstituted with limited numbers of T cells from lethal challenge with IHD-J-Luc vaccinia virus. J Virol 2015; 89:3295-307. [PMID: 25589648 DOI: 10.1128/jvi.03340-14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Protection from lethality by postchallenge administration of brincidofovir (BCV, CMX001) was studied in normal and immune-deficient (nude, nu/nu) BALB/c mice infected with vaccinia virus (VACV). Whole-body bioluminescence imaging was used to record total fluxes in the nasal cavity, lungs, spleen, and liver and to enumerate pox lesions on tails of mice infected via the intranasal route with 10(5) PFU of recombinant IHD-J-Luc VACV expressing luciferase. Areas under the flux curve (AUCs) were calculated for individual mice to assess viral loads. A three-dose regimen of 20 mg/kg BCV administered every 48 h starting either on day 1 or day 2 postchallenge protected 100% of mice. Initiating BCV treatment earlier was more efficient in reducing viral loads and in providing protection from pox lesion development. All BCV-treated mice that survived challenge were also protected from rechallenge with IHD-J-Luc or WRvFire VACV without additional treatment. In immune-deficient mice, BCV protected animals from lethality and reduced viral loads while animals were on the drug. Viral recrudescence occurred within 4 to 9 days, and mice succumbed ∼10 to 20 days after treatment termination. Nude mice reconstituted with 10(5) T cells prior to challenge with 10(4) PFU of IHD-J-Luc and treated with BCV postchallenge survived the infection, cleared the virus from all organs, and survived rechallenge with 10(5) PFU of IHD-J-Luc VACV without additional BCV treatment. Together, these data suggest that BCV protects immunocompetent and partially T cell-reconstituted immune-deficient mice from lethality, reduces viral dissemination in organs, prevents pox lesion development, and permits generation of VACV-specific memory. IMPORTANCE Mass vaccination is the primary element of the public health response to a smallpox outbreak. In addition to vaccination, however, antiviral drugs are required for individuals with uncertain exposure status to smallpox or for whom vaccination is contraindicated. Whole-body bioluminescence imaging was used to study the effect of brincidofovir (BCV) in normal and immune-deficient (nu/nu) mice infected with vaccinia virus, a model of smallpox. Postchallenge administration of 20 mg/kg BCV rescued normal and immune-deficient mice partially reconstituted with T cells from lethality and significantly reduced viral loads in organs. All BCV-treated mice that survived infection were protected from rechallenge without additional treatment. In immune-deficient mice, BCV extended survival. The data show that BCV controls viral replication at the site of challenge and reduces viral dissemination to internal organs, thus providing a shield for the developing adaptive immunity that clears the host of virus and builds virus-specific immunological memory.
Collapse
|
112
|
Falendysz EA, Londoño-Navas AM, Meteyer CU, Pussini N, Lopera JG, Osorio JE, Rocke TE. Evaluation of monkeypox virus infection of black-tailed prairie dogs (Cynomys ludovicianus) using in vivo bioluminescent imaging. J Wildl Dis 2014; 50:524-36. [PMID: 24779460 PMCID: PMC4636010 DOI: 10.7589/2013-07-171] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Monkeypox (MPX) is a re-emerging zoonotic disease that is endemic in Central and West Africa, where it can cause a smallpox-like disease in humans. Despite many epidemiologic and field investigations of MPX, no definitive reservoir species has been identified. Using recombinant viruses expressing the firefly luciferase (luc) gene, we previously demonstrated the suitability of in vivo bioluminescent imaging (BLI) to study the pathogenesis of MPX in animal models. Here, we evaluated BLI as a novel approach for tracking MPX virus infection in black-tailed prairie dogs (Cynomys ludovicianus). Prairie dogs were affected during a multistate outbreak of MPX in the US in 2003 and have since been used as an animal model of this disease. Our BLI results were compared with PCR and virus isolation from tissues collected postmortem. Virus was easily detected and quantified in skin and superficial tissues by BLI before and during clinical phases, as well as in subclinical secondary cases, but was not reliably detected in deep tissues such as the lung. Although there are limitations to viral detection in larger wild rodent species, BLI can enhance the use of prairie dogs as an animal model of MPX and can be used for the study of infection, disease progression, and transmission in potential wild rodent reservoirs.
Collapse
Affiliation(s)
- Elizabeth A Falendysz
- 1 US Geological Survey-National Wildlife Health Center, 6006 Schroeder Road, Madison, Wisconsin 53711, USA
| | | | | | | | | | | | | |
Collapse
|
113
|
Nagata N, Saijo M, Kataoka M, Ami Y, Suzaki Y, Sato Y, Iwata-Yoshikawa N, Ogata M, Kurane I, Morikawa S, Sata T, Hasegawa H. Pathogenesis of fulminant monkeypox with bacterial sepsis after experimental infection with West African monkeypox virus in a cynomolgus monkey. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:4359-70. [PMID: 25120821 PMCID: PMC4129056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 07/01/2014] [Indexed: 06/03/2023]
Abstract
The pathogenesis of severe human monkeypox, which causes systemic and fulminant infections, is not clear. This study presents a case repot of fulminant monkeypox with bacterial sepsis after experimental infection with monkeypox virus in a cynomolgus monkey (Macaca fascicularis). In our previous study (Saijo et al., 2009, J Gen Virol), two cynomolgus monkeys became moribund after experimental infection with monkeypox virus Liberia strain, West African strain. One exhibited typical monkeypox-related papulovesicular lesions. The other monkey presented fulminant clinical symptoms with a characteristic flat red rash similar to that found in smallpox, which is associated with extremely high fatality rates. In this study, we found that the monkey with flat red rash had high levels of viremia and neutropenia, as well as high plasma levels of pro-inflammatory cytokines and chemokines compared with the other monkey. Monkeypox virus replicates in epithelial cells and macrophages in various organs. Sepsis due to Gram-positive cocci was confirmed histopathologically in the monkey with flat red rash. The lack of inflammatory response in the lesion suggested that the monkey with sepsis experienced strong immune suppression during the viral infection. The neutropenia and excessive inflammatory cytokine responses indicate that neutrophils play key roles in the pathogenesis of systemic and fulminant human monkeypox virus infections with sepsis.
Collapse
Affiliation(s)
- Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Masayuki Saijo
- Department of Virology 1, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Michiyo Kataoka
- Department of Pathology, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Yasushi Ami
- Department of Division of Experimental Animals Research, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Yuriko Suzaki
- Department of Division of Experimental Animals Research, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Yuko Sato
- Department of Pathology, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Naoko Iwata-Yoshikawa
- Department of Pathology, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Momoko Ogata
- Department of Virology 1, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Ichiro Kurane
- Department of Virology 1, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Shigeru Morikawa
- Department of Veterinary Science, National Institute of Infectious Diseases1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Tetsutaro Sata
- Department of Pathology, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Hideki Hasegawa
- Department of Pathology, National Institute of Infectious Diseases4-7-1 Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| |
Collapse
|
114
|
|
115
|
Song H, Sidney J, Wiseman RW, Josleyn N, Cohen M, Blaney JE, Jahrling PB, Sette A. Characterizing monkeypox virus specific CD8+ T cell epitopes in rhesus macaques. Virology 2013; 447:181-6. [PMID: 24210113 PMCID: PMC4771384 DOI: 10.1016/j.virol.2013.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 08/13/2013] [Accepted: 09/03/2013] [Indexed: 11/18/2022]
Abstract
To characterize T cell epitopes in monkeypox virus (MPXV) infected rhesus macaques, we utilized IFNγ Elispot assay to screen 400 predicted peptides from 20MPXV proteins. Two peptides from the F8L protein, an analog of E9L protein in vaccinia, were found to elicit CD8+ T cell responses. Prediction and in vitro MHC binding analyses suggest that one is restricted by Mamu-A1(⁎)001 and another by Mamu-A1(⁎)002. The Mamu-A1(⁎)002 epitope is completely identical in all reported sequences for variola, vaccinia, cowpox and MPXV. The Mamu-A1(⁎)001 epitope is conserved in MPXV and vaccinia, and has one residue substitution (V6>I) in some cowpox sequences and all variola sequences. Given CD8+ T-cell epitopes from E9L were also identified in humans and mice, our data suggested that F8L/E9L may be a dominant pox viral protein for CD8+ T cell responses, and may be considered as a target when designing vaccines that target pox-specific T cell responses.
Collapse
Affiliation(s)
- Haifeng Song
- Integrated Research Facility, NIAID/NIH, Frederick, MD 21702, USA.
| | | | | | | | | | | | | | | |
Collapse
|
116
|
Song H, Josleyn N, Janosko K, Skinner J, Reeves RK, Cohen M, Jett C, Johnson R, Blaney JE, Bollinger L, Jennings G, Jahrling PB. Monkeypox virus infection of rhesus macaques induces massive expansion of natural killer cells but suppresses natural killer cell functions. PLoS One 2013; 8:e77804. [PMID: 24147080 PMCID: PMC3798392 DOI: 10.1371/journal.pone.0077804] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 09/05/2013] [Indexed: 12/16/2022] Open
Abstract
Natural killer (NK) cells play critical roles in innate immunity and in bridging innate and adaptive immune responses against viral infection. However, the response of NK cells to monkeypox virus (MPXV) infection is not well characterized. In this intravenous challenge study of MPXV infection in rhesus macaques (Macaca mulatta), we analyzed blood and lymph node NK cell changes in absolute cell numbers, cell proliferation, chemokine receptor expression, and cellular functions. Our results showed that the absolute number of total NK cells in the blood increased in response to MPXV infection at a magnitude of 23-fold, manifested by increases in CD56+, CD16+, CD16-CD56- double negative, and CD16+CD56+ double positive NK cell subsets. Similarly, the frequency and NK cell numbers in the lymph nodes also largely increased with the total NK cell number increasing 46.1-fold. NK cells both in the blood and lymph nodes massively proliferated in response to MPXV infection as measured by Ki67 expression. Chemokine receptor analysis revealed reduced expression of CXCR3, CCR7, and CCR6 on NK cells at early time points (days 2 and 4 after virus inoculation), followed by an increased expression of CXCR3 and CCR5 at later time points (days 7-8) of infection. In addition, MPXV infection impaired NK cell degranulation and ablated secretion of interferon-γ and tumor necrosis factor-α. Our data suggest a dynamic model by which NK cells respond to MPXV infection of rhesus macaques. Upon virus infection, NK cells proliferated robustly, resulting in massive increases in NK cell numbers. However, the migrating capacity of NK cells to tissues at early time points might be reduced, and the functions of cytotoxicity and cytokine secretion were largely compromised. Collectively, the data may explain, at least partially, the pathogenesis of MPXV infection in rhesus macaques.
Collapse
Affiliation(s)
- Haifeng Song
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
- * E-mail:
| | - Nicole Josleyn
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Krisztina Janosko
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Jeff Skinner
- Computational Biology Section, Bioinformatics and Computational Biosciences Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland, United States of America
| | - R. Keith Reeves
- New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America
| | - Melanie Cohen
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Catherine Jett
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Reed Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland, United States of America
| | - Joseph E. Blaney
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland, United States of America
| | - Laura Bollinger
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Gerald Jennings
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Peter B. Jahrling
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland, United States of America
| |
Collapse
|
117
|
Keckler MS, Reynolds MG, Damon IK, Karem KL. The effects of post-exposure smallpox vaccination on clinical disease presentation: addressing the data gaps between historical epidemiology and modern surrogate model data. Vaccine 2013; 31:5192-201. [PMID: 23994378 DOI: 10.1016/j.vaccine.2013.08.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Revised: 08/05/2013] [Accepted: 08/13/2013] [Indexed: 11/28/2022]
Abstract
Decades after public health interventions - including pre- and post-exposure vaccination - were used to eradicate smallpox, zoonotic orthopoxvirus outbreaks and the potential threat of a release of variola virus remain public health concerns. Routine prophylactic smallpox vaccination of the public ceased worldwide in 1980, and the adverse event rate associated with the currently licensed live vaccinia virus vaccine makes reinstatement of policies recommending routine pre-exposure vaccination unlikely in the absence of an orthopoxvirus outbreak. Consequently, licensing of safer vaccines and therapeutics that can be used post-orthopoxvirus exposure is necessary to protect the global population from these threats. Variola virus is a solely human pathogen that does not naturally infect any other known animal species. Therefore, the use of surrogate viruses in animal models of orthopoxvirus infection is important for the development of novel vaccines and therapeutics. Major complications involved with the use of surrogate models include both the absence of a model that accurately mimics all aspects of human smallpox disease and a lack of reproducibility across model species. These complications limit our ability to model post-exposure vaccination with newer vaccines for application to human orthopoxvirus outbreaks. This review seeks to (1) summarize conclusions about the efficacy of post-exposure smallpox vaccination from historic epidemiological reports and modern animal studies; (2) identify data gaps in these studies; and (3) summarize the clinical features of orthopoxvirus-associated infections in various animal models to identify those models that are most useful for post-exposure vaccination studies. The ultimate purpose of this review is to provide observations and comments regarding available model systems and data gaps for use in improving post-exposure medical countermeasures against orthopoxviruses.
Collapse
Affiliation(s)
- M Shannon Keckler
- Centers for Disease Control and Prevention, Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, United States.
| | | | | | | |
Collapse
|
118
|
Hatch GJ, Graham VA, Bewley KR, Tree JA, Dennis M, Taylor I, Funnell SGP, Bate SR, Steeds K, Tipton T, Bean T, Hudson L, Atkinson DJ, McLuckie G, Charlwood M, Roberts ADG, Vipond J. Assessment of the protective effect of Imvamune and Acam2000 vaccines against aerosolized monkeypox virus in cynomolgus macaques. J Virol 2013; 87:7805-15. [PMID: 23658452 PMCID: PMC3700201 DOI: 10.1128/jvi.03481-12] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 04/30/2013] [Indexed: 12/17/2022] Open
Abstract
To support the licensure of a new and safer vaccine to protect people against smallpox, a monkeypox model of infection in cynomolgus macaques, which simulates smallpox in humans, was used to evaluate two vaccines, Acam2000 and Imvamune, for protection against disease. Animals vaccinated with a single immunization of Imvamune were not protected completely from severe and/or lethal infection, whereas those receiving either a prime and boost of Imvamune or a single immunization with Acam2000 were protected completely. Additional parameters, including clinical observations, radiographs, viral load in blood, throat swabs, and selected tissues, vaccinia virus-specific antibody responses, immunophenotyping, extracellular cytokine levels, and histopathology were assessed. There was no significant difference (P > 0.05) between the levels of neutralizing antibody in animals vaccinated with a single immunization of Acam2000 (132 U/ml) and the prime-boost Imvamune regime (69 U/ml) prior to challenge with monkeypox virus. After challenge, there was evidence of viral excretion from the throats of 2 of 6 animals in the prime-boost Imvamune group, whereas there was no confirmation of excreted live virus in the Acam2000 group. This evaluation of different human smallpox vaccines in cynomolgus macaques helps to provide information about optimal vaccine strategies in the absence of human challenge studies.
Collapse
Affiliation(s)
- Graham J Hatch
- Microbiological Services, Public Health England, Salisbury, Wiltshire, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
119
|
Song H, Janosko K, Johnson RF, Qin J, Josleyn N, Jett C, Byrum R, Claire MS, Dyall J, Blaney JE, Jennings G, Jahrling PB. Poxvirus antigen staining of immune cells as a biomarker to predict disease outcome in monkeypox and cowpox virus infection in non-human primates. PLoS One 2013; 8:e60533. [PMID: 23577120 PMCID: PMC3618230 DOI: 10.1371/journal.pone.0060533] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 02/27/2013] [Indexed: 11/19/2022] Open
Abstract
Infection of non-human primates (NHPs) such as rhesus and cynomolgus macaques with monkeypox virus (MPXV) or cowpox virus (CPXV) serve as models to study poxvirus pathogenesis and to evaluate vaccines and anti-orthopox therapeutics. Intravenous inoculation of macaques with high dose of MPXV (>1-2×10(7) PFU) or CPXV (>10(2) PFU) results in 80% to 100% mortality and 66 to 100% mortality respectively. Here we report that NHPs with positive detection of poxvirus antigens in immune cells by flow cytometric staining, especially in monocytes and granulocytes succumbed to virus infection and that early positive pox staining is a strong predictor for lethality. Samples from four independent studies were analyzed. Eighteen NHPs from three different experiments were inoculated with two different MPXV strains at lethal doses. Ten NHPs displayed positive pox-staining and all 10 NHPs reached moribund endpoint. In contrast, none of the three NHPs that survived anticipated lethal virus dose showed apparent virus staining in the monocytes and granulocytes. In addition, three NHPs that were challenged with a lethal dose of MPXV and received cidofovir treatment were pox-antigen negative and all three NHPs survived. Furthermore, data from a CPXV study also demonstrated that 6/9 NHPs were pox-antigen staining positive and all 6 NHPs reached euthanasia endpoint, while the three survivors were pox-antigen staining negative. Thus, we conclude that monitoring pox-antigen staining in immune cells can be used as a biomarker to predict the prognosis of virus infection. Future studies should focus on the mechanisms and implications of the pox-infection of immune cells and the correlation between pox-antigen detection in immune cells and disease progression in human poxviral infection.
Collapse
Affiliation(s)
- Haifeng Song
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Krisztina Janosko
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Reed F. Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jing Qin
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Nicole Josleyn
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Catherine Jett
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Russell Byrum
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Marisa St. Claire
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Julie Dyall
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Joseph E. Blaney
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Gerald Jennings
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Peter B. Jahrling
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
120
|
Hutson CL, Gallardo-Romero N, Carroll DS, Clemmons C, Salzer JS, Nagy T, Hughes CM, Olson VA, Karem KL, Damon IK. Transmissibility of the monkeypox virus clades via respiratory transmission: investigation using the prairie dog-monkeypox virus challenge system. PLoS One 2013; 8:e55488. [PMID: 23408990 PMCID: PMC3567100 DOI: 10.1371/journal.pone.0055488] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 12/27/2012] [Indexed: 11/24/2022] Open
Abstract
Monkeypox virus (MPXV) is endemic within Africa where it sporadically is reported to cause outbreaks of human disease. In 2003, an outbreak of human MPXV occurred in the US after the importation of infected African rodents. Since the eradication of smallpox (caused by an orthopoxvirus (OPXV) related to MPXV) and cessation of routine smallpox vaccination (with the live OPXV vaccinia), there is an increasing population of people susceptible to OPXV diseases. Previous studies have shown that the prairie dog MPXV model is a functional animal model for the study of systemic human OPXV illness. Studies with this model have demonstrated that infected animals are able to transmit the virus to naive animals through multiple routes of exposure causing subsequent infection, but were not able to prove that infected animals could transmit the virus exclusively via the respiratory route. Herein we used the model system to evaluate the hypothesis that the Congo Basin clade of MPXV is more easily transmitted, via respiratory route, than the West African clade. Using a small number of test animals, we show that transmission of viruses from each of the MPXV clade was minimal via respiratory transmission. However, transmissibility of the Congo Basin clade was slightly greater than West African MXPV clade (16.7% and 0% respectively). Based on these findings, respiratory transmission appears to be less efficient than those of previous studies assessing contact as a mechanism of transmission within the prairie dog MPXV animal model.
Collapse
Affiliation(s)
- Christina L. Hutson
- Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Diseases, Atlanta, Georgia, United States of America
- Department of Pathology, College of Veterinary Medicine, The University of Georgia, Athens, Georgia, United States of America
| | - Nadia Gallardo-Romero
- Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Diseases, Atlanta, Georgia, United States of America
| | - Darin S. Carroll
- Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Diseases, Atlanta, Georgia, United States of America
| | - Cody Clemmons
- Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Diseases, Atlanta, Georgia, United States of America
| | - Johanna S. Salzer
- Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Diseases, Atlanta, Georgia, United States of America
- Program in Population Biology, Ecology and Evolution, Emory University, Atlanta, Georgia, United States of America
| | - Tamas Nagy
- Department of Pathology, College of Veterinary Medicine, The University of Georgia, Athens, Georgia, United States of America
| | - Christine M. Hughes
- Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Diseases, Atlanta, Georgia, United States of America
| | - Victoria A. Olson
- Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Diseases, Atlanta, Georgia, United States of America
| | - Kevin L. Karem
- Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Diseases, Atlanta, Georgia, United States of America
| | - Inger K. Damon
- Division of High-Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Diseases, Atlanta, Georgia, United States of America
| |
Collapse
|
121
|
Parker S, Buller RM. A review of experimental and natural infections of animals with monkeypox virus between 1958 and 2012. Future Virol 2013; 8:129-157. [PMID: 23626656 DOI: 10.2217/fvl.12.130] [Citation(s) in RCA: 161] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Monkeypox virus (MPXV) was discovered in 1958 during an outbreak in an animal facility in Copenhagen, Denmark. Since its discovery, MPXV has revealed a propensity to infect and induce disease in a large number of animals within the mammalia class from pan-geographical locations. This finding has impeded the elucidation of the natural host, although the strongest candidates are African squirrels and/or other rodents. Experimentally, MPXV can infect animals via a variety of multiple different inoculation routes; however, the natural route of transmission is unknown and is likely to be somewhat species specific. In this review we have attempted to compile and discuss all published articles that describe experimental or natural infections with MPXV, dating from the initial discovery of the virus through to the year 2012. We further discuss the comparative disease courses and pathologies of the host species.
Collapse
Affiliation(s)
- Scott Parker
- Department of Molecular Microbiology & Immunology, Saint Louis University School of Medicine, 1100 S. Grand Blvd, Saint Louis, MO 63104, USA
| | | |
Collapse
|
122
|
Townsend MB, Keckler MS, Patel N, Davies DH, Felgner P, Damon IK, Karem KL. Humoral immunity to smallpox vaccines and monkeypox virus challenge: proteomic assessment and clinical correlations. J Virol 2013; 87:900-11. [PMID: 23135728 PMCID: PMC3554095 DOI: 10.1128/jvi.02089-12] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 10/25/2012] [Indexed: 11/20/2022] Open
Abstract
Despite the eradication of smallpox, orthopoxviruses (OPV) remain public health concerns. Efforts to develop new therapeutics and vaccines for smallpox continue through their evaluation in animal models despite limited understanding of the specific correlates of protective immunity. Recent monkeypox virus challenge studies have established the black-tailed prairie dog (Cynomys ludovicianus) as a model of human systemic OPV infections. In this study, we assess the induction of humoral immunity in humans and prairie dogs receiving Dryvax, Acam2000, or Imvamune vaccine and characterize the proteomic profile of immune recognition using enzyme-linked immunosorbent assays (ELISA), neutralization assays, and protein microarrays. We confirm anticipated similarities of antigenic protein targets of smallpox vaccine-induced responses in humans and prairie dogs and identify several differences. Subsequent monkeypox virus intranasal infection of vaccinated prairie dogs resulted in a significant boost in humoral immunity characterized by a shift in reactivity of increased intensity to a broader range of OPV proteins. This work provides evidence of similarities between the vaccine responses in prairie dogs and humans that enhance the value of the prairie dog model system as an OPV vaccination model and offers novel findings that form a framework for examining the humoral immune response induced by systemic orthopoxvirus infection.
Collapse
Affiliation(s)
- M B Townsend
- Centers for Disease Control and Prevention, Division of High Consequence Pathogens and Pathology, Poxvirus and Rabies Branch, Atlanta, GA, USA.
| | | | | | | | | | | | | |
Collapse
|
123
|
Cann JA, Jahrling PB, Hensley LE, Wahl-Jensen V. Comparative pathology of smallpox and monkeypox in man and macaques. J Comp Pathol 2013; 148:6-21. [PMID: 22884034 PMCID: PMC3498598 DOI: 10.1016/j.jcpa.2012.06.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 06/01/2012] [Accepted: 06/19/2012] [Indexed: 11/21/2022]
Abstract
In the three decades since the eradication of smallpox and cessation of routine vaccination, the collective memory of the devastating epidemics caused by this orthopoxvirus has waned, and the human population has become increasingly susceptible to a disease that remains high on the list of possible bioterrorism agents. Research using surrogate orthopoxviruses in their natural hosts, as well as limited variola virus research in animal models, continues worldwide; however, interpretation of findings is often limited by our relative lack of knowledge about the naturally occurring disease. For modern comparative pathologists, many of whom have no first-hand knowledge of naturally occurring smallpox, this work provides a contemporary review of this historical disease, as well as discussion of how it compares with human monkeypox and the corresponding diseases in macaques.
Collapse
Affiliation(s)
- J A Cann
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA.
| | | | | | | |
Collapse
|
124
|
Abstract
As the threat of exposure to emerging and reemerging viruses within a naive population increases, it is vital that the basic mechanisms of pathogenesis and immune response be thoroughly investigated. By using animal models in this endeavor, the response to viruses can be studied in a more natural context to identify novel drug targets, and assess the efficacy and safety of new products. This is especially true in the advent of the Food and Drug Administration's animal rule. Although no one animal model is able to recapitulate all the aspects of human disease, understanding the current limitations allows for a more targeted experimental design. Important facets to be considered before an animal study are the route of challenge, species of animals, biomarkers of disease, and a humane endpoint. This chapter covers the current animal models for medically important human viruses, and demonstrates where the gaps in knowledge exist.
Collapse
|
125
|
Nectin-4-dependent measles virus spread to the cynomolgus monkey tracheal epithelium: role of infected immune cells infiltrating the lamina propria. J Virol 2012; 87:2526-34. [PMID: 23255790 DOI: 10.1128/jvi.03037-12] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After the contagion measles virus (MV) crosses the respiratory epithelium within myeloid cells that express the primary receptor signaling lymphocytic activation molecule (SLAM), it replicates briskly in SLAM-expressing cells in lymphatic organs. Later, the infection spreads to epithelia expressing nectin-4, an adherens junction protein expressed preferentially in the trachea, but how it gets there is not understood. To characterize the mechanisms of spread, we infected groups of 5 or 6 cynomolgus monkeys (Macaca fascicularis) with either a wild-type MV or its "N4-blind" derivative, which is unable to enter nectin-4-expressing cells because of the targeted mutation of two hemagglutinin residues. As expected, both viruses caused similar levels of immunosuppression, as monitored by reductions in white blood cell counts and lymphocyte proliferation activity. However, monkeys infected with the N4-blind MV cleared infection more rapidly. Wild-type virus-infected monkeys secreted virus, while marginal virus titers were detected in tracheal lavage fluid cells of N4-blind MV-infected hosts. Analyses of tracheal rings obtained at necropsy (day 12) documented widespread infection of individual cells or small cell clusters in the subepithelial lamina propria of monkeys infected with either virus. However, only wild-type MV spread to the epithelium, forming numerous infectious centers comprised of many contiguous columnar cells. Infected CD11c(+) myeloid (macrophage or dendritic) cells were frequently observed in the lamina propria below epithelial infectious centers. Thus, MV may use myeloid cells as vehicles not only immediately after contagion but also to infect epithelia of tissues expressing nectin-4, including the trachea.
Collapse
|
126
|
Barnewall RE, Fisher DA, Robertson AB, Vales PA, Knostman KA, Bigger JE. Inhalational monkeypox virus infection in cynomolgus macaques. Front Cell Infect Microbiol 2012; 2:117. [PMID: 23061051 PMCID: PMC3443748 DOI: 10.3389/fcimb.2012.00117] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 08/20/2012] [Indexed: 11/14/2022] Open
Abstract
An inhalation exposure system was characterized to deliver aerosolized monkeypox virus (MPXV), and a non-human primate (NHP) inhalation monkeypox model was developed in cynomolgus macaques. A head-only aerosol exposure system was characterized, and two sampling methods were evaluated: liquid impingement via an impinger and impaction via a gelatin filter. The aerosol concentrations obtained with the gelatin filter and impinger were virtually identical, indicating that either method is acceptable for sampling aerosols containing MPXV. The mass median aerodynamic diameter (MMAD) for individual aerosol tests in the aerosol system characterization and the NHP study ranged from 1.08 to 1.15 μm, indicating that the aerosol particles were of a sufficient size to reach the alveoli. Six cynomolgus macaques (four male and two female) were used on study. The animals were aerosol exposed with MPXV and received doses between 2.51 × 10(4) to 9.28 × 10(5) plaque forming units (PFUs) inhaled. Four of the six animals died or were euthanized due to their moribund conditions. Both animals that received the lowest exposure doses survived to the end of the observation period. The inhalation LD(50) was determined to be approximately 7.8 × 10(4) pfu inhaled. These data demonstrate that an inhalation MPXV infection model has been developed in the cynomolgus macaque with disease course and lethal dose similar to previously published data.
Collapse
|
127
|
Rivetti AV, Guedes MIMC, Rehfeld IS, Oliveira TML, Matos ACD, Abrahão JS, Kroon EG, Lobato ZIP. Bovine vaccinia, a systemic infection: evidence of fecal shedding, viremia and detection in lymphoid organs. Vet Microbiol 2012; 162:103-11. [PMID: 23021861 DOI: 10.1016/j.vetmic.2012.09.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 08/30/2012] [Accepted: 09/05/2012] [Indexed: 11/15/2022]
Abstract
Bovine vaccinia (BV) is a zoonosis caused by Vaccinia virus (VACV) that affects dairy cattle and milkers, causing economic losses and impacting animal and human health. Based on the clinical presentation, BV appears to be a localized disease, with lesions restricted to the skin of affected individuals. However, there are no studies on the pathogenesis of the disease in cows to determine if there is a systemic spread of the virus and if there are different ways of VACV shedding. The objective of this work was to study if there is a systemic spread of VACV in experimentally infected cows and to study the kinetics of VACV circulation in the blood and shedding in the feces of these animals. To this end, eight crossbred lactating cows were used. Three teats of each cow were inoculated with the GP2V strain of VACV. All animals were monitored daily, and blood and fecal samples were collected for 67 days post-infection (dpi). After this period, four of these previously infected cows were immunosuppressed using dexamethasone. Viral DNA was continuously detected and quantified in the blood and feces of these animals in an intermittent way, even after the resolution of the lesions. At slaughter, tissues were collected, and viral DNA was detected and quantified in the mesenteric and retromammary lymph nodes, ileum, spleen and liver. The detection of VACV DNA in the feces for a longer period (67 dpi) and in the lymphatic organs provides new evidence about VACV elimination and suggests that BV could be a systemic infection with a chronic course and viral shedding through the feces.
Collapse
Affiliation(s)
- Anselmo V Rivetti
- Departamento de Medicina Veterinária Preventiva, Escola de Veterinária, Universidade Federal de Minas Gerais (UFMG) - Av. Antônio Carlos 6627, Caixa Postal 567, Campus da UFMG, CEP 30123-970, Belo Horizonte, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
128
|
Earl PL, Americo JL, Moss B. Lethal monkeypox virus infection of CAST/EiJ mice is associated with a deficient gamma interferon response. J Virol 2012; 86:9105-12. [PMID: 22696658 PMCID: PMC3416162 DOI: 10.1128/jvi.00162-12] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 06/05/2012] [Indexed: 11/20/2022] Open
Abstract
Monkeypox virus (MPXV) is endemic in Africa, where it causes disease in humans resembling smallpox. A recent importation of MPXV-infected animals into the United States raises the possibility of global spread. Rodents comprise the major reservoir of MPXV, and a variety of such animals, even those native to North America, are susceptible. In contrast, common inbred strains of mice, including BALB/c and C57BL/6, are greatly resistant to MPXV. However, several inbred strains of mice derived from wild mice, including CAST/EiJ, exhibit morbidity and mortality at relatively low inoculums of MPXV. Elucidating the basis for the susceptibility of CAST/EiJ mice could contribute to an understanding of MPXV pathogenicity and host defense mechanisms and enhance the value of this mouse strain as a model system for evaluation of therapeutics and vaccines. Here we compared virus dissemination and induced cytokine production in CAST/EiJ mice to those in the resistant BALB/c strain. Following intranasal infection, robust virus replication occurred in the lungs of both strains, although a relatively higher inoculum was required for BALB/c. However, while spread to other internal organs was rapid and efficient in CAST/EiJ mice, the virus was largely restricted to the lungs in BALB/c mice. Gamma interferon (IFN-γ) and CCL5 were induced in lungs of BALB/c mice concomitant with virus replication but not in CAST/EiJ mice. The importance of IFN-γ in protection against MPXV disease was demonstrated by the intranasal administration of the mouse cytokine to CAST/EiJ mice and the resulting protection against MPXV. Furthermore, C57BL/6 mice with inactivation of the IFN-γ gene or the IFN-γ receptor gene exhibited enhanced sensitivity to MPXV.
Collapse
Affiliation(s)
- Patricia L Earl
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | |
Collapse
|
129
|
Bhunu CP, Mushayabasa S, Hyman JM. Modelling HIV/AIDS and monkeypox co-infection. APPLIED MATHEMATICS AND COMPUTATION 2012; 218:9504-9518. [PMID: 36345302 PMCID: PMC9629068 DOI: 10.1016/j.amc.2012.03.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
During the last two decades, reports on emerging human monkeypox outbreaks in Africa and North America have reminded us that beside the eradicated smallpox there are other pox viruses that have great potential to harm people. A deterministic model for the co-infection of HIV/AIDS and monkeypox is formulated and analysed. The endemic equilibria are shown to be locally and globally asymptotically stable using the Centre Manifold theory and the Lyapunov function approach, respectively. Analysis of the basic reproduction numbers and numerical simulations suggest that an increase in the number of monkeypox in the animal species results in an increase of the number of people having monkeypox. Threshold conditions that determine the competitive outcomes of the two diseases are provided. Furthermore, numerical simulations using a set of reasonable parameter values support the claim that HIV infection greatly enhances monkeypox infection and vice versa.
Collapse
Affiliation(s)
- C P Bhunu
- Department of Applied Mathematics, Modelling Biomedical Systems Research Group, National University of Science and Technology, P. O. Box 939, Ascot, Bulawayo, Zimbabwe
- Department of Veterinary Medicine, University of Cambridge, Cambridge, CB3 OES, UK
| | - S Mushayabasa
- Department of Applied Mathematics, Modelling Biomedical Systems Research Group, National University of Science and Technology, P. O. Box 939, Ascot, Bulawayo, Zimbabwe
| | - J M Hyman
- Mathematics Department, Tulane University, New Orleans, LA 70118, USA
| |
Collapse
|
130
|
Breithaupt A, Kalthoff D, Deutskens F, König P, Hoffmann B, Beer M, Meyer H, Teifke JP. Clinical course and pathology in rats (Rattus norvegicus) after experimental cowpox virus infection by percutaneous and intranasal application. Vet Pathol 2012; 49:941-9. [PMID: 22411549 DOI: 10.1177/0300985812439077] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Recently, several cases of human cowpox virus (CPXV) infections were reported in France and Germany, which had been acquired through close contact with infected pet rats. The animals exhibited respiratory signs or skin lesions and died shortly after purchase. After natural infection of white rats with CPXV in the USSR in 1978, a peracute pulmonary form, a milder dermal form, and a mixed form exhibiting features of both have been described. To the best of the authors' knowledge, 3 experimental cowpox virus infection studies using rats have been performed to date; however, neither results of histomorphological examinations nor immunohistochemical analyses have yet been reported in rats after experimental infections. To investigate the impact of the infection route on the clinical course, the development of lesions, and tropism, rats were infected intradermally, intranasally, or by a combination of both routes. The authors found a correlation between clinical manifestation, pathology, and infection routes. Intradermal and contact exposure yielded a mild dermal form, characterized by the development of vesiculopustular dermatitis. In contrast, intranasally infected animals died peracutely, showing severe dyspnea. Occasionally, a combination of the dermal and the respiratory form occurred after intranasal infection. Immunohistochemically, CPXV antigen was detected in the epithelial and mesenchymal cells of the upper respiratory tract and affected skin lesions and rarely in mesenchymal cells of lymph nodes. This is the first histomorphological and immunohistochemical analysis of CPXV in rats after experimental infection.
Collapse
Affiliation(s)
- A Breithaupt
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Südufer 10, 17493 Greifswald-Insel, Riems, Germany
| | | | | | | | | | | | | | | |
Collapse
|
131
|
Embury-Hyatt C, Babiuk S, Manning L, Ganske S, Bowden T, Boyle D, Copps J. Pathology and viral antigen distribution following experimental infection of sheep and goats with capripoxvirus. J Comp Pathol 2012; 146:106-15. [PMID: 22297076 PMCID: PMC9528194 DOI: 10.1016/j.jcpa.2011.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 10/27/2011] [Accepted: 12/02/2011] [Indexed: 12/02/2022]
Abstract
Current understanding of capripoxvirus pathogenesis is limited since there have been no detailed studies examining cell tropism at well-defined intervals following infection. We undertook time-course studies in sheep and goats following inoculation of sheeppox or goatpox viruses in their respective homologous hosts, and examined tissues by light microscopy. A monoclonal antibody generated to a sheeppox virus core protein was used for immunohistochemical detection of viral antigen in tissue sections. Lesions and virus antigen were observed consistently in the skin, lung and lymph nodes. Antigen was detected at 6 and 8 days post inoculation for skin and lung, respectively, within cells which appeared to be of monocyte/macrophage lineage. In sheep skin capripoxvirus immunoreactivity was detected within previously unreported large multinucleated cells. In the lung, double immunolabelling detected the simultaneous expression of capripoxvirus antigen and cytokeratin indicating the presence of virus within pneumocytes. Lung double immunolabelling also detected the expression of capripoxvirus antigen in CD68(+) cells, confirming the presence of viral antigen within macrophages. Based on early detection of infected macrophages, dissemination of virus within the host and localization to tissues likely occurred through cells of the monocyte/macrophage lineage. Histological findings revealed similarities with both monkeypox and smallpox, thus capripoxvirus infection in sheep and goats may represent useful models with which to study strategies for poxvirus-specific virus vaccine concepts and therapeutics.
Collapse
Affiliation(s)
- C. Embury-Hyatt
- National Centre for Foreign Animal Disease, 1015 Arlington Street, Winnipeg, Manitoba, Canada,Correspondence to: C. Embury-Hyatt
| | - S. Babiuk
- National Centre for Foreign Animal Disease, 1015 Arlington Street, Winnipeg, Manitoba, Canada,University of Manitoba, Department of Immunology, Basic Medical Sciences Building, 730 William Avenue, Winnipeg, Manitoba, Canada
| | - L. Manning
- National Centre for Foreign Animal Disease, 1015 Arlington Street, Winnipeg, Manitoba, Canada
| | - S. Ganske
- National Centre for Foreign Animal Disease, 1015 Arlington Street, Winnipeg, Manitoba, Canada
| | - T.R. Bowden
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia
| | - D.B. Boyle
- CSIRO Livestock Industries, Australian Animal Health Laboratory, Private Bag 24, Geelong, Victoria 3220, Australia
| | - J. Copps
- National Centre for Foreign Animal Disease, 1015 Arlington Street, Winnipeg, Manitoba, Canada
| |
Collapse
|
132
|
|
133
|
Dyall J, Johnson RF, Chen DY, Huzella L, Ragland DR, Mollura DJ, Byrum R, Reba RC, Jennings G, Jahrling PB, Blaney JE, Paragas J. Evaluation of monkeypox disease progression by molecular imaging. J Infect Dis 2011; 204:1902-11. [PMID: 22013221 PMCID: PMC3209815 DOI: 10.1093/infdis/jir663] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 08/09/2011] [Indexed: 11/13/2022] Open
Abstract
Infection of nonhuman primates (NHPs) with monkeypox virus (MPXV) is currently being developed as an animal model of variola infection in humans. We used positron emission tomography and computed tomography (PET/CT) to identify inflammatory patterns as predictors for the outcome of MPXV disease in NHPs. Two NHPs were sublethally inoculated by the intravenous (IV) or intrabronchial (IB) routes and imaged sequentially using fluorine-18 fluorodeoxyglucose ((18)FDG) uptake as a nonspecific marker of inflammation/immune activation. Inflammation was observed in the lungs of IB-infected NHPs, and bilobular involvement was associated with morbidity. Lymphadenopathy and immune activation in the axillary lymph nodes were evident in IV- and IB-infected NHPs. Interestingly, the surviving NHPs had significant (18)FDG uptake in the axillary lymph nodes at the time of MPXV challenge with no clinical signs of illness, suggesting an association between preexisting immune activation and survival. Molecular imaging identified patterns of inflammation/immune activation that may allow risk assessment of monkeypox disease.
Collapse
Affiliation(s)
- Julie Dyall
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases
| | - Reed F. Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases
| | - Dar-Yeong Chen
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases
| | - Louis Huzella
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases
| | - Dan R. Ragland
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases
| | - Daniel J. Mollura
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Russell Byrum
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases
| | - Richard C. Reba
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Gerald Jennings
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases
| | - Peter B. Jahrling
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases
| | - Joseph E. Blaney
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases
| | - Jason Paragas
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases
| |
Collapse
|
134
|
Wahl-Jensen V, Cann JA, Rubins KH, Huggins JW, Fisher RW, Johnson AJ, de Kok-Mercado F, Larsen T, Raymond JL, Hensley LE, Jahrling PB. Progression of pathogenic events in cynomolgus macaques infected with variola virus. PLoS One 2011; 6:e24832. [PMID: 21998632 PMCID: PMC3188545 DOI: 10.1371/journal.pone.0024832] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 08/20/2011] [Indexed: 11/18/2022] Open
Abstract
Smallpox, caused by variola virus (VARV), is a devastating human disease that affected millions worldwide until the virus was eradicated in the 1970 s. Subsequent cessation of vaccination has resulted in an immunologically naive human population that would be at risk should VARV be used as an agent of bioterrorism. The development of antivirals and improved vaccines to counter this threat would be facilitated by the development of animal models using authentic VARV. Towards this end, cynomolgus macaques were identified as adequate hosts for VARV, developing ordinary or hemorrhagic smallpox in a dose-dependent fashion. To further refine this model, we performed a serial sampling study on macaques exposed to doses of VARV strain Harper calibrated to induce ordinary or hemorrhagic disease. Several key differences were noted between these models. In the ordinary smallpox model, lymphoid and myeloid hyperplasias were consistently found whereas lymphocytolysis and hematopoietic necrosis developed in hemorrhagic smallpox. Viral antigen accumulation, as assessed immunohistochemically, was mild and transient in the ordinary smallpox model. In contrast, in the hemorrhagic model antigen distribution was widespread and included tissues and cells not involved in the ordinary model. Hemorrhagic smallpox developed only in the presence of secondary bacterial infections – an observation also commonly noted in historical reports of human smallpox. Together, our results support the macaque model as an excellent surrogate for human smallpox in terms of disease onset, acute disease course, and gross and histopathological lesions.
Collapse
Affiliation(s)
- Victoria Wahl-Jensen
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Jennifer A. Cann
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Kathleen H. Rubins
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - John W. Huggins
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Robert W. Fisher
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Anthony J. Johnson
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Fabian de Kok-Mercado
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Thomas Larsen
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Jo Lynne Raymond
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Lisa E. Hensley
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Peter B. Jahrling
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
135
|
Goff A, Mucker E, Raymond J, Fisher R, Bray M, Hensley L, Paragas J. Infection of cynomolgus macaques with a recombinant monkeypox virus encoding green fluorescent protein. Arch Virol 2011; 156:1877-81. [PMID: 21814864 DOI: 10.1007/s00705-011-1065-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 07/01/2011] [Indexed: 11/26/2022]
Abstract
Monkeypox virus (MPXV) causes a vesiculopustular rash illness resembling smallpox in humans and produces a similar disease in nonhuman primates. To enhance the ability of researchers to study experimental MPXV infections, we inserted a gene encoding green fluorescent protein (GFP) into Monkeypox virus Zaire-79. Wild-type and MPXV-GFP replicated with similar kinetics in cell culture and caused a similar disease when injected intravenously into cynomolgus macaques. In MPXV-GFP-infected animals, examination under fluorescent light facilitated the identification of skin lesions during disease development and internal sites of replication at necropsy. MPXV-GFP could improve the quantitative assessment of antiviral therapy and vaccine efficacy.
Collapse
Affiliation(s)
- Arthur Goff
- Virology Division, Viral Therapeutics Branch, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702-5011, USA.
| | | | | | | | | | | | | |
Collapse
|
136
|
Smith AL, St Claire M, Yellayi S, Bollinger L, Jahrling PB, Paragas J, Blaney JE, Johnson RF. Intrabronchial inoculation of cynomolgus macaques with cowpox virus. J Gen Virol 2011; 93:159-164. [PMID: 21940414 DOI: 10.1099/vir.0.036905-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The public health threat of orthopoxviruses from bioterrorist attacks has prompted researchers to develop suitable animal models for increasing our understanding of viral pathogenesis and evaluation of medical countermeasures (MCMs) in compliance with the FDA Animal Efficacy Rule. We present an accessible intrabronchial cowpox virus (CPXV) model that can be evaluated under biosafety level-2 laboratory conditions. In this dose-ranging study, utilizing cynomolgus macaques, signs of typical orthopoxvirus disease were observed with the lymphoid organs, liver, skin (generally mild) and respiratory tract as target tissues. Clinical and histopathological evaluation suggests that intrabronchial CPXV recapitulated many of the features of monkeypox and variola virus, the causative agent of smallpox, infections in cynomolgus macaque models. These similarities suggest that CPXV infection in non-human primates should be pursued further as an alternative model of smallpox. Further development of the CPXV primate model, unimpeded by select agent and biocontainment restrictions, should facilitate the development of MCMs for smallpox.
Collapse
Affiliation(s)
- Alvin L Smith
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marisa St Claire
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Srikanth Yellayi
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Laura Bollinger
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Peter B Jahrling
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA.,Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jason Paragas
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD 21702, USA
| | - Joseph E Blaney
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Reed F Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
137
|
Estep RD, Messaoudi I, O'Connor MA, Li H, Sprague J, Barron A, Engelmann F, Yen B, Powers MF, Jones JM, Robinson BA, Orzechowska BU, Manoharan M, Legasse A, Planer S, Wilk J, Axthelm MK, Wong SW. Deletion of the monkeypox virus inhibitor of complement enzymes locus impacts the adaptive immune response to monkeypox virus in a nonhuman primate model of infection. J Virol 2011; 85:9527-42. [PMID: 21752919 PMCID: PMC3165757 DOI: 10.1128/jvi.00199-11] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 06/26/2011] [Indexed: 01/13/2023] Open
Abstract
Monkeypox virus (MPXV) is an orthopoxvirus closely related to variola virus, the causative agent of smallpox. Human MPXV infection results in a disease that is similar to smallpox and can also be fatal. Two clades of MPXV have been identified, with viruses of the central African clade displaying more pathogenic properties than those within the west African clade. The monkeypox inhibitor of complement enzymes (MOPICE), which is not expressed by viruses of the west African clade, has been hypothesized to be a main virulence factor responsible for increased pathogenic properties of central African strains of MPXV. To gain a better understanding of the role of MOPICE during MPXV-mediated disease, we compared the host adaptive immune response and disease severity following intrabronchial infection with MPXV-Zaire (n = 4), or a recombinant MPXV-Zaire (n = 4) lacking expression of MOPICE in rhesus macaques (RM). Data presented here demonstrate that infection of RM with MPXV leads to significant viral replication in the peripheral blood and lungs and results in the induction of a robust and sustained adaptive immune response against the virus. More importantly, we show that the loss of MOPICE expression results in enhanced viral replication in vivo, as well as a dampened adaptive immune response against MPXV. Taken together, these findings suggest that MOPICE modulates the anti-MPXV immune response and that this protein is not the sole virulence factor of the central African clade of MPXV.
Collapse
Affiliation(s)
- Ryan D. Estep
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon
| | - Ilhem Messaoudi
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon
| | - Megan A. O'Connor
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon
| | - Helen Li
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon
| | - Jerald Sprague
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon
| | - Alexander Barron
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon
| | - Flora Engelmann
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon
| | - Bonnie Yen
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon
| | - Michael F. Powers
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon
| | - John M. Jones
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon
| | - Bridget A. Robinson
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon
| | - Beata U. Orzechowska
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon
| | - Minsha Manoharan
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon
| | - Alfred Legasse
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon
| | - Shannon Planer
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon
| | - Jennifer Wilk
- Division of Animal Resources, Oregon National Primate Research Center, Beaverton, Oregon
| | - Michael K. Axthelm
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon
| | - Scott W. Wong
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon
| |
Collapse
|
138
|
Johnson RF, Yellayi S, Cann JA, Johnson A, Smith AL, Paragas J, Jahrling PB, Blaney JE. Cowpox virus infection of cynomolgus macaques as a model of hemorrhagic smallpox. Virology 2011; 418:102-12. [PMID: 21840027 DOI: 10.1016/j.virol.2011.07.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 07/15/2011] [Accepted: 07/18/2011] [Indexed: 01/27/2023]
Abstract
Hemorrhagic smallpox was a rare but severe manifestation of variola virus infection that resulted in nearly 100% mortality. Here we describe intravenous (IV) inoculation of cowpox virus Brighton Red strain in cynomolgus macaques (Macaca fascicularis) which resulted in disease similar in presentation to hemorrhagic smallpox in humans. IV inoculation of macaques resulted in a uniformly lethal disease within 12 days post-inoculation in two independent experiments. Clinical observations and hematological and histopathological findings support hemorrhagic disease. Cowpox virus replicated to high levels in blood (8.0-9.0 log(10) gene copies/mL) and tissues including lymph nodes, thymus, spleen, bone marrow, and lungs. This unique model of hemorrhagic orthopoxvirus infection provides an accessible means to further study orthopoxvirus pathogenesis and to identify virus-specific and nonspecific therapies. Such studies will serve to complement the existing nonhuman primate models of more classical poxviral disease.
Collapse
Affiliation(s)
- Reed F Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
139
|
Mätz-Rensing K, Stahl-Hennig C, Kramski M, Pauli G, Ellerbrok H, Kaup FJ. The pathology of experimental poxvirus infection in common marmosets (Callithrix jacchus): further characterization of a new primate model for orthopoxvirus infections. J Comp Pathol 2011; 146:230-42. [PMID: 21783202 DOI: 10.1016/j.jcpa.2011.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 05/18/2011] [Accepted: 06/06/2011] [Indexed: 10/18/2022]
Abstract
Zoonotic orthopoxvirus (OPV) can induce severe disease in man and the virus has potential for use in bioterrorism. New vaccines and therapeutics against OPV infections must be tested in animal models. The aim of this study was to characterize the clinical course and pathology of a new OPV isolate, calpox virus, which is infectious in marmosets. Infection experiments were performed with 28 common marmosets (Callithrix jacchus) exposed to different challenge doses of calpox virus by the intravenous, oropharyngeal and intranasal (IN) routes. The median marmoset IN infectious dose corresponded to 8.3 × 10(2)plaque forming units of calpox virus. Infected animals developed reproducible clinical signs and died within 4-15 days post infection. Characteristic pox-like lesions developed in affected organs, particularly in the skin, mucous membranes, lymph nodes, liver and spleen. Calpox virus disease progression and pathological findings in the common marmoset appear to be consistent with lethal OPV infections in man and in other non-human primate (NHP) models. IN inoculation with low virus doses mimics the natural route of the human variola virus infection. Thus, the marmoset model of calpox virus infection can be considered to be relevant to investigation of the mechanisms of OPV pathogenesis and pathology and for the evaluation of new vaccines and antiviral therapies.
Collapse
|
140
|
Grosenbach DW, Jordan R, Hruby DE. Development of the small-molecule antiviral ST-246 as a smallpox therapeutic. Future Virol 2011; 6:653-671. [PMID: 21837250 DOI: 10.2217/fvl.11.27] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Naturally occurring smallpox has been eradicated, yet it remains as one of the highest priority pathogens due to its potential as a biological weapon. The majority of the US population would be vulnerable in a smallpox outbreak. SIGA Technologies, Inc. has responded to the call of the US government to develop and supply to the Strategic National Stockpile a smallpox antiviral to be deployed in the event of a smallpox outbreak. ST-246(®) (tecovirimat) was initially identified via a high-throughput screen in 2002, and in the ensuing years, our drug-development activities have spanned in vitro analysis, preclinical safety, pharmacokinetics and efficacy testing (all according to the 'animal rule'). Additionally, SIGA has conducted Phase I and II clinical trials to evaluate the safety, tolerability and pharmacokinetics of ST-246, bringing us to our current late stage of clinical development. This article reviews the need for a smallpox therapeutic and our experience in developing ST-246, and provides perspective on the role of a smallpox antiviral during a smallpox public health emergency.
Collapse
Affiliation(s)
- Douglas W Grosenbach
- SIGA Technologies, Inc., 4575 SW Research Way, Suite 230, Corvallis, OR 97333, USA
| | | | | |
Collapse
|
141
|
Goff AJ, Chapman J, Foster C, Wlazlowski C, Shamblin J, Lin K, Kreiselmeier N, Mucker E, Paragas J, Lawler J, Hensley L. A novel respiratory model of infection with monkeypox virus in cynomolgus macaques. J Virol 2011; 85:4898-909. [PMID: 21389129 PMCID: PMC3126178 DOI: 10.1128/jvi.02525-10] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Accepted: 02/28/2011] [Indexed: 11/20/2022] Open
Abstract
Variola, the causative agent of smallpox, and the related monkeypox virus are both select agents that, if purposefully released, would cause public panic and social disruption. For this reason research continues in the areas of animal model and therapeutic development. Orthopoxviruses show a widely varying degree of host specificity, making development of accurate animal models difficult. In this paper, we demonstrate a novel respiratory infection technique that resulted in "classic" orthopox disease in nonhuman primates and takes the field of research one step closer to a better animal model.
Collapse
Affiliation(s)
- Arthur J Goff
- United States Army Medical Research Institute of Infectious Diseases, 1425 Porter St., Virology Division, Viral Therapeutics Branch, 903Q, Fort Detrick, MD 21702-5011, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Gordon SN, Cecchinato V, Andresen V, Heraud JM, Hryniewicz A, Parks RW, Venzon D, Chung HK, Karpova T, McNally J, Silvera P, Reimann KA, Matsui H, Kanehara T, Shinmura Y, Yokote H, Franchini G. Smallpox vaccine safety is dependent on T cells and not B cells. J Infect Dis 2011; 203:1043-53. [PMID: 21450994 PMCID: PMC3068024 DOI: 10.1093/infdis/jiq162] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Accepted: 11/03/2010] [Indexed: 11/13/2022] Open
Abstract
The licensed smallpox vaccine, ACAM2000, is a cell culture derivative of Dryvax. Both ACAM2000 and Dryvax are administered by skin scarification and can cause progressive vaccinia, with skin lesions that disseminate to distal sites. We have investigated the immunologic basis of the containment of vaccinia in the skin with the goal to identify safer vaccines for smallpox. Macaques were depleted systemically of T or B cells and vaccinated with either Dryvax or an attenuated vaccinia vaccine, LC16m8. B cell depletion did not affect the size of skin lesions induced by either vaccine. However, while depletion of both CD4(+) and CD8(+) T cells had no adverse effects on LC16m8-vaccinated animals, it caused progressive vaccinia in macaques immunized with Dryvax. As both Dryvax and LC16m8 vaccines protect healthy macaques from a lethal monkeypox intravenous challenge, our data identify LC16m8 as a safer and effective alternative to ACAM2000 and Dryvax vaccines for immunocompromised individuals.
Collapse
Affiliation(s)
| | | | | | - Jean-Michel Heraud
- World Health Organization-National Influenza Laboratory, Institut Pasteur de Madagascar, Antananarivo, Madagascar
| | | | | | | | | | - Tatiana Karpova
- Fluorescence Imaging Facility, Laboratory of Receptor Biology, Gene Expression and Metabolism
| | - James McNally
- National Cancer Institute, Bethesda, and Southern Research Institute, Frederick
| | - Peter Silvera
- National Cancer Institute, Bethesda, and Southern Research Institute, Frederick
| | - Keith A. Reimann
- Division of Viral Pathogenesis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Hajime Matsui
- The Chemo-Sero-Therapeutic Research Institute (KAKETSUKEN), Kumamoto, Japan
| | - Tomomi Kanehara
- The Chemo-Sero-Therapeutic Research Institute (KAKETSUKEN), Kumamoto, Japan
| | - Yasuhiko Shinmura
- The Chemo-Sero-Therapeutic Research Institute (KAKETSUKEN), Kumamoto, Japan
| | - Hiroyuki Yokote
- The Chemo-Sero-Therapeutic Research Institute (KAKETSUKEN), Kumamoto, Japan
| | | |
Collapse
|
143
|
Johnson RF, Dyall J, Ragland DR, Huzella L, Byrum R, Jett C, St Claire M, Smith AL, Paragas J, Blaney JE, Jahrling PB. Comparative analysis of monkeypox virus infection of cynomolgus macaques by the intravenous or intrabronchial inoculation route. J Virol 2011; 85:2112-25. [PMID: 21147922 PMCID: PMC3067809 DOI: 10.1128/jvi.01931-10] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Accepted: 11/17/2010] [Indexed: 11/20/2022] Open
Abstract
Monkeypox virus (MPXV) infection has recently expanded in geographic distribution and can be fatal in up to 10% of cases. The intravenous (i.v.) inoculation of nonhuman primates (NHPs) results in an accelerated fulminant disease course compared to that of naturally occurring MPXV infection in humans. Alternative routes of inoculation are being investigated to define an NHP model of infection that more closely resembles natural disease progression. Our goal was to determine if the intrabronchial (i.b.) exposure of NHPs to MPXV results in a systemic disease that better resembles the progression of human MPXV infection. Here, we compared the disease course following an i.v. or i.b. inoculation of NHPs with 10-fold serial doses of MPXV Zaire. Classical pox-like disease was observed in NHPs administered a high virus dose by either route. Several key events were delayed in the highest doses tested of the i.b. model compared to the timing of the i.v. model, including the onset of fever, lesion appearance, peak viremia, viral shedding in nasal and oral swabs, peak cytokine levels, and time to reach endpoint criteria. Virus distribution across 19 tissues was largely unaffected by the inoculation route at the highest doses tested. The NHPs inoculated by the i.b. route developed a viral pneumonia that likely exacerbated disease progression. Based on the observations of the delayed onset of clinical and virological parameters and endpoint criteria that may more closely resemble those of human MPXV infection, the i.b. MPXV model should be considered for the further investigation of viral pathogenesis and countermeasures.
Collapse
Affiliation(s)
- Reed F Johnson
- National Institutes of Health, NIAID/EVPS, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Rubins KH, Hensley LE, Relman DA, Brown PO. Stunned silence: gene expression programs in human cells infected with monkeypox or vaccinia virus. PLoS One 2011; 6:e15615. [PMID: 21267444 PMCID: PMC3022624 DOI: 10.1371/journal.pone.0015615] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 11/12/2010] [Indexed: 12/20/2022] Open
Abstract
Poxviruses use an arsenal of molecular weapons to evade detection and disarm host immune responses. We used DNA microarrays to investigate the gene expression responses to infection by monkeypox virus (MPV), an emerging human pathogen, and Vaccinia virus (VAC), a widely used model and vaccine organism, in primary human macrophages, primary human fibroblasts and HeLa cells. Even as the overwhelmingly infected cells approached their demise, with extensive cytopathic changes, their gene expression programs appeared almost oblivious to poxvirus infection. Although killed (gamma-irradiated) MPV potently induced a transcriptional program characteristic of the interferon response, no such response was observed during infection with either live MPV or VAC. Moreover, while the gene expression response of infected cells to stimulation with ionomycin plus phorbol 12-myristate 13-acetate (PMA), or poly (I-C) was largely unimpaired by infection with MPV, a cluster of pro-inflammatory genes were a notable exception. Poly(I-C) induction of genes involved in alerting the innate immune system to the infectious threat, including TNF-alpha, IL-1 alpha and beta, CCL5 and IL-6, were suppressed by infection with live MPV. Thus, MPV selectively inhibits expression of genes with critical roles in cell-signaling pathways that activate innate immune responses, as part of its strategy for stealthy infection.
Collapse
Affiliation(s)
- Kathleen H. Rubins
- Departments of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
| | - Lisa E. Hensley
- United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - David A. Relman
- Departments of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Patrick O. Brown
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
145
|
Abstract
The eradication of smallpox, one of the great triumphs of medicine, was accomplished through the prophylactic administration of live vaccinia virus, a comparatively benign relative of variola virus, the causative agent of smallpox. Nevertheless, recent fears that variola virus may be used as a biological weapon together with the present susceptibility of unimmunized populations have spurred the development of new-generation vaccines that are safer than the original and can be produced by modern methods. Predicting the efficacy of such vaccines in the absence of human smallpox, however, depends on understanding the correlates of protection. This review outlines the biology of poxviruses with particular relevance to vaccine development, describes protein targets of humoral and cellular immunity, compares animal models of orthopoxvirus disease with human smallpox, and considers the status of second- and third-generation smallpox vaccines.
Collapse
Affiliation(s)
- Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-3210, USA.
| |
Collapse
|
146
|
Brown JN, Estep RD, Lopez-Ferrer D, Brewer HM, Clauss TR, Manes NP, O'Connor M, Li H, Adkins JN, Wong SW, Smith RD. Characterization of macaque pulmonary fluid proteome during monkeypox infection: dynamics of host response. Mol Cell Proteomics 2010; 9:2760-71. [PMID: 20736407 PMCID: PMC3101861 DOI: 10.1074/mcp.m110.001875] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 08/13/2010] [Indexed: 01/13/2023] Open
Abstract
Understanding viral pathogenesis is challenging because of confounding factors, including nonabrasive access to infected tissues and high abundance of inflammatory mediators that may mask mechanistic details. In diseases such as influenza and smallpox where the primary cause of mortality results from complications in the lung, the characterization of lung fluid offers a unique opportunity to study host-pathogen interactions with minimal effect on infected animals. This investigation characterizes the global proteome response in the pulmonary fluid, bronchoalveolar lavage fluid, of macaques during upper respiratory infection by monkeypox virus (MPXV), a close relative of the causative agent of smallpox, variola virus. These results are compared and contrasted against infections by vaccinia virus (VV), a low pathogenic relative of MPXV, and with extracellular fluid from MPXV-infected HeLa cells. To identify changes in the pulmonary protein compartment, macaque lung fluid was sampled twice prior to infection, serving as base line, and up to six times following intrabronchial infection with either MPXV or VV. Increased expression of inflammatory proteins was observed in response to both viruses. Although the increased expression resolved for a subset of proteins, such as C-reactive protein, S100A8, and S100A9, high expression levels persisted for other proteins, including vitamin D-binding protein and fibrinogen γ. Structural and metabolic proteins were substantially decreased in lung fluid exclusively during MPXV and not VV infection. Decreases in structural and metabolic proteins were similarly observed in the extracellular fluid of MPXV-infected HeLa cells. Results from this study suggest that the host inflammatory response may not be the only facilitator of viral pathogenesis, but rather maintaining pulmonary structural integrity could be a key factor influencing disease progression and mortality.
Collapse
Affiliation(s)
- Joseph N. Brown
- From the ‡Fundamental Science Division and Environmental Molecular Science Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352 and
| | - Ryan D. Estep
- §Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon 97006
| | - Daniel Lopez-Ferrer
- From the ‡Fundamental Science Division and Environmental Molecular Science Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352 and
| | - Heather M. Brewer
- From the ‡Fundamental Science Division and Environmental Molecular Science Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352 and
| | - Theresa R. Clauss
- From the ‡Fundamental Science Division and Environmental Molecular Science Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352 and
| | - Nathan P. Manes
- From the ‡Fundamental Science Division and Environmental Molecular Science Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352 and
| | - Megan O'Connor
- §Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon 97006
| | - Helen Li
- §Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon 97006
| | - Joshua N. Adkins
- From the ‡Fundamental Science Division and Environmental Molecular Science Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352 and
| | - Scott W. Wong
- §Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon 97006
| | - Richard D. Smith
- From the ‡Fundamental Science Division and Environmental Molecular Science Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352 and
| |
Collapse
|
147
|
Jordan R, Leeds JM, Tyavanagimatt S, Hruby DE. Development of ST-246® for Treatment of Poxvirus Infections. Viruses 2010; 2:2409-2435. [PMID: 21994624 PMCID: PMC3185582 DOI: 10.3390/v2112409] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2010] [Revised: 10/26/2010] [Accepted: 10/26/2010] [Indexed: 12/26/2022] Open
Abstract
ST-246 (Tecovirimat) is a small synthetic antiviral compound being developed to treat pathogenic orthopoxvirus infections of humans. The compound was discovered as part of a high throughput screen designed to identify inhibitors of vaccinia virus-induced cytopathic effects. The antiviral activity is specific for orthopoxviruses and the compound does not inhibit the replication of other RNA- and DNA-containing viruses or inhibit cell proliferation at concentrations of compound that are antiviral. ST-246 targets vaccinia virus p37, a viral protein required for envelopment and secretion of extracellular forms of virus. The compound is orally bioavailable and protects multiple animal species from lethal orthopoxvirus challenge. Preclinical safety pharmacology studies in mice and non-human primates indicate that ST-246 is readily absorbed by the oral route and well tolerated with the no observable adverse effect level (NOAEL) in mice measured at 2000 mg/kg and the no observable effect level (NOEL) in non-human primates measured at 300 mg/kg. Drug substance and drug product processes have been developed and commercial scale batches have been produced using Good Manufacturing Processes (GMP). Human phase I clinical trials have shown that ST-246 is safe and well tolerated in healthy human volunteers. Based on the results of the clinical evaluation, once a day dosing should provide plasma drug exposure in the range predicted to be antiviral based on data from efficacy studies in animal models of orthopoxvirus disease. These data support the use of ST-246 as a therapeutic to treat pathogenic orthopoxvirus infections of humans.
Collapse
Affiliation(s)
- Robert Jordan
- SIGA Technologies, 4575 SW Research Way, Corvallis, OR 97333, USA; E-Mails: (J.M.L); (S.T.); (D.E.H.)
| | - Janet M. Leeds
- SIGA Technologies, 4575 SW Research Way, Corvallis, OR 97333, USA; E-Mails: (J.M.L); (S.T.); (D.E.H.)
| | | | - Dennis E. Hruby
- SIGA Technologies, 4575 SW Research Way, Corvallis, OR 97333, USA; E-Mails: (J.M.L); (S.T.); (D.E.H.)
| |
Collapse
|
148
|
Nalca A, Livingston VA, Garza NL, Zumbrun EE, Frick OM, Chapman JL, Hartings JM. Experimental infection of cynomolgus macaques (Macaca fascicularis) with aerosolized monkeypox virus. PLoS One 2010; 5:e12880. [PMID: 20862223 PMCID: PMC2942837 DOI: 10.1371/journal.pone.0012880] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 08/11/2010] [Indexed: 11/18/2022] Open
Abstract
Monkeypox virus (MPXV) infection in humans results in clinical symptoms very similar to ordinary smallpox. Aerosol is a route of secondary transmission for monkeypox, and a primary route of smallpox transmission in humans. Therefore, an animal model for aerosol exposure to MPXV is needed to test medical countermeasures. To characterize the pathogenesis in cynomolgus macaques (Macaca fascicularis), groups of macaques were exposed to four different doses of aerosolized MPXV. Blood was collected the day before, and every other day after exposure and assessed for complete blood count (CBC), clinical chemistry analysis, and quantitative PCR. Macaques showed mild anorexia, depression, and fever on day 6 post-exposure. Lymphadenopathy, which differentiates monkeypox from smallpox, was observed in exposed macaques around day 6 post-exposure. CBC and clinical chemistries showed abnormalities similar to human monkeypox cases. Whole blood and throat swab viral loads peaked around day 10, and in survivors, gradually decreased until day 28 post-exposure. Survival was not dose dependent. As such, doses of 4 × 10(4) PFU, 1 × 10(5) PFU, or 1 × 10(6) PFU resulted in lethality for 70% of the animals, whereas a dose of 4 × 10(5) PFU resulted in 85% lethality. Overall, cynomolgus macaques exposed to aerosolized MPXV develop a clinical disease that resembles that of human monkeypox. These findings provide a strong foundation for the use of aerosolized MPXV exposure of cynomolgus macaques as an animal model to test medical countermeasures against orthopoxviruses.
Collapse
Affiliation(s)
- Aysegul Nalca
- Center for Aerobiological Sciences, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA.
| | | | | | | | | | | | | |
Collapse
|
149
|
Meseda CA, Weir JP. Third-generation smallpox vaccines: challenges in the absence of clinical smallpox. Future Microbiol 2010; 5:1367-82. [DOI: 10.2217/fmb.10.98] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Smallpox, a disease caused by variola virus, is estimated to have killed hundreds of millions to billions of people before it was certified as eradicated in 1980. However, there has been renewed interest in smallpox vaccine development due in part to zoonotic poxvirus infections and the possibility of a re-emergence of smallpox, as well as the fact that first-generation smallpox vaccines are associated with relatively rare, but severe, adverse reactions in some vaccinees. An understanding of the immune mechanisms of vaccine protection and the use of suitable animal models for vaccine efficacy assessment are paramount to the development of safer and effective smallpox vaccines. This article focuses on studies aimed at understanding the immune responses elicited by vaccinia virus and the various animal models that can be used to evaluate smallpox vaccine efficacy. Harnessing this information is necessary to assess the effectiveness and potential usefulness of new-generation smallpox vaccines.
Collapse
Affiliation(s)
| | - Jerry P Weir
- Division of Viral Products, Center for Biologics Evaluation & Research, USFDA, 1401 Rockville Pike, HFM-457, Rockville, MD 20852, USA
| |
Collapse
|
150
|
Chapman JL, Nichols DK, Martinez MJ, Raymond JW. Animal models of orthopoxvirus infection. Vet Pathol 2010; 47:852-70. [PMID: 20682806 DOI: 10.1177/0300985810378649] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Smallpox was one of the most devastating diseases known to humanity. Although smallpox was eradicated through a historically successful vaccination campaign, there is concern in the global community that either Variola virus (VARV), the causative agent of smallpox, or another species of Orthopoxvirus could be used as agents of bioterrorism. Therefore, development of countermeasures to Orthopoxvirus infection is a crucial focus in biodefense research, and these efforts rely on the use of various animal models. Smallpox typically presented as a generalized pustular rash with 30 to 40% mortality, and although smallpox-like syndromes can be induced in cynomolgus macaques with VARV, research with this virus is highly restricted; therefore, animal models with other orthopoxviruses have been investigated. Monkeypox virus causes a generalized vesiculopustular rash in rhesus and cynomolgus macaques and induces fatal systemic disease in several rodent species. Ectromelia virus has been extensively studied in mice as a model of orthopoxviral infection in its natural host. Intranasal inoculation of mice with some strains of vaccinia virus produces fatal bronchopneumonia, as does aerosol or intranasal inoculation of mice with cowpox virus. Rabbitpox virus causes pneumonia and fatal systemic infections in rabbits and can be naturally transmitted between rabbits by an aerosol route similar to that of VARV in humans. No single animal model recapitulates all known aspects of human Orthopoxvirus infections, and each model has its advantages and disadvantages. This article provides a brief review of the Orthopoxvirus diseases of humans and the key pathologic features of animal models of Orthopoxvirus infections.
Collapse
Affiliation(s)
- J L Chapman
- DVM, Major, US Army, US Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, MD 21702, USA.
| | | | | | | |
Collapse
|