151
|
Mutations in the E2 Glycoprotein and the 3' Untranslated Region Enhance Chikungunya Virus Virulence in Mice. J Virol 2017; 91:JVI.00816-17. [PMID: 28747508 DOI: 10.1128/jvi.00816-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/21/2017] [Indexed: 01/22/2023] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that causes debilitating musculoskeletal pain and inflammation and can persist for months to years after acute infection. Although studies of humans and experimentally infected animals suggest that CHIKV infection persists in musculoskeletal tissues, the mechanisms for this remain poorly understood. To evaluate this further, we isolated CHIKV from the serum of persistently infected Rag1 -/- mice at day 28. When inoculated into naive wild-type (WT) mice, this persistently circulating CHIKV strain displayed a capacity for earlier dissemination and greater pathogenicity than the parental virus. Sequence analysis revealed a nonsynonymous mutation in the E2 glycoprotein (E2 K200R) and a deletion within the 3' untranslated region (3'-UTR). The introduction of these changes into the parental virus conferred enhanced virulence in mice, although primary tropism for musculoskeletal tissues was maintained. The E2 K200R mutation was largely responsible for enhanced viral dissemination and pathogenicity, although these effects were augmented by the 3'-UTR deletion. Finally, studies with Irf3/Irf7 -/- and Ifnar1 -/- mice suggest that the E2 K200R mutation enhances viral dissemination from the site of inoculation independently of interferon regulatory factor 3 (IRF3)-, IRF7-, and IFNAR1-mediated responses. As our findings reveal viral determinants of CHIKV dissemination and pathogenicity, their further study should help to elucidate host-virus interactions that determine acute and chronic CHIKV infection.IMPORTANCE CHIKV is a globally spreading, mosquito-transmitted virus that causes debilitating acute and chronic musculoskeletal disease in humans. The viral genetic determinants that dictate the severity of acute and chronic diseases are not understood. To improve our understanding of CHIKV pathogenesis, we evaluated a CHIKV strain isolated from the serum of chronically infected immunocompromised mice. Sequence analysis of this persistent CHIKV strain identified two mutations, an amino acid change in the E2 viral attachment protein and a deletion within the 3'-UTR of the viral genome. We identified roles for these mutations in the enhancement of viral dissemination from the inoculation site and in disease severity. These data improve our understanding of the viral determinants of CHIKV pathogenesis and adaptive changes that occur during viral persistence.
Collapse
|
152
|
Zivcec M, Guerrero LIW, Albariño CG, Bergeron É, Nichol ST, Spiropoulou CF. Identification of broadly neutralizing monoclonal antibodies against Crimean-Congo hemorrhagic fever virus. Antiviral Res 2017; 146:112-120. [PMID: 28842265 DOI: 10.1016/j.antiviral.2017.08.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/17/2017] [Accepted: 08/22/2017] [Indexed: 02/07/2023]
Abstract
Despite the serious public health impact of Crimean-Congo hemorrhagic fever (CCHF), the efficacy of antivirals targeting the causative agent, CCHF virus (CCHFV), remains debatable. Neutralizing monoclonal antibodies (MAbs) targeting the CCHFV glycoprotein Gc have been reported to protect mice against challenge with the prototype CCHFV strain, IbAr10200. However, due to extensive sequence diversity of CCHFV glycoproteins, it is unknown whether these MAbs neutralize other CCHFV strains. We initially used a CCHF virus-like particle (VLP) system to generate 11 VLP moieties, each possessing a glycoprotein from a genetically diverse CCHFV strain isolated in either Africa, Asia, the Middle East, or southeastern Europe. We used these VLPs in biosafety level 2 conditions to efficiently screen MAb cross-neutralization potency. Of the 16 MAbs tested, 3 (8A1, 11E7, and 30F7) demonstrated cross-neutralization activity with most CCHF VLPs, with 8A1 neutralizing all VLPs tested. Although binding studies suggest that none of the MAbs compete for the same epitope, combining 11E7, 30F7, or both 11E7 and 30F7 with 8A1 had no additive effect on increasing neutralization in this system. To confirm our findings from the VLP system, the 3 MAbs capable of strain cross-neutralization were confirmed to effectively neutralize 5 diverse CCHFV strains in vitro. Passaging CCHFV strains in the presence of sub-neutralizing concentrations of MAbs did not generate escape mutants resistant to subsequent neutralization. This study demonstrates the utility of the VLP system for screening neutralizing MAbs against multiple CCHFV strains, and provides the first evidence that a single MAb can effectively neutralize a number of diverse CCHFV strains in vitro, which may lead to development of future CCHF therapeutics.
Collapse
Affiliation(s)
- Marko Zivcec
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Lisa I W Guerrero
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - César G Albariño
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Éric Bergeron
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Stuart T Nichol
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| |
Collapse
|
153
|
Saborío Galo S, González K, Téllez Y, García N, Pérez L, Gresh L, Harris E, Balmaseda Á. Development of in-house serological methods for diagnosis and surveillance of chikungunya. Rev Panam Salud Publica 2017; 41:e56. [PMID: 28902269 PMCID: PMC5638038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 06/23/2016] [Indexed: 10/13/2023] Open
Abstract
OBJECTIVE To develop and evaluate serological methods for chikungunya diagnosis and research in Nicaragua. METHODS Two IgM ELISA capture systems (MAC-ELISA) for diagnosis of acute chikungunya virus (CHIKV) infections, and two Inhibition ELISA Methods (IEM) to measure total antibodies against CHIKV were developed using monoclonal antibodies (mAbs) and hyperimmune serum at the National Virology Laboratory of Nicaragua in 2014-2015. The sensitivity, specificity, predictive values, and agreement of the MAC-ELISAs were obtained by comparing the results of 198 samples (116 positive; 82 negative) with the Centers for Disease Control and Prevention's IgM ELISA (Atlanta, Georgia, United States; CDC-MAC-ELISA). For clinical evaluation of the four serological techniques, 260 paired acute and convalescent phase serum samples of suspected chikungunya cases were used. RESULTS All four assays were standardized by determining the optimal concentrations of the different reagents. Processing times were substantially reduced compared to the CDC-MAC-ELISA. For the MAC-ELISA systems, a sensitivity of 96.6% and 97.4%, and a specificity of 98.8% and 91.5% were obtained using mAb and hyperimmune serum, respectively, compared with the CDC method. Clinical evaluation of the four serological techniques versus the CDC real-time RT-PCR assay resulted in a sensitivity of 95.7% and a specificity of 88.8%-95.9%. CONCLUSION Two MAC-ELISA and two IEM systems were standardized, demonstrating very good quality for chikungunya diagnosis and research demands. This will achieve more efficient epidemiological surveillance in Nicaragua, the first country in Central America to produce its own reagents for serological diagnosis of CHIKV. The methods evaluated here can be applied in other countries and will contribute to sustainable diagnostic systems to combat the disease.
Collapse
Affiliation(s)
- Saira Saborío Galo
- National Virology Laboratory, Centro Nacional de Diagnóstico y ReferenciaMinistry of HealthManaguaNicaraguaNational Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua.
| | - Karla González
- National Virology Laboratory, Centro Nacional de Diagnóstico y ReferenciaMinistry of HealthManaguaNicaraguaNational Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua.
| | - Yolanda Téllez
- National Virology Laboratory, Centro Nacional de Diagnóstico y ReferenciaMinistry of HealthManaguaNicaraguaNational Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua.
| | - Nadezna García
- National Virology Laboratory, Centro Nacional de Diagnóstico y ReferenciaMinistry of HealthManaguaNicaraguaNational Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua.
| | - Leonel Pérez
- National Virology Laboratory, Centro Nacional de Diagnóstico y ReferenciaMinistry of HealthManaguaNicaraguaNational Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua.
| | - Lionel Gresh
- Sustainable Sciences InstituteSustainable Sciences InstituteManaguaNicaraguaSustainable Sciences Institute, Managua, Nicaragua.
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public HealthUniversity of CaliforniaBerkeleyCaliforniaUnited States of AmericaDivision of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, California, United States of America.
| | - Ángel Balmaseda
- National Virology Laboratory, Centro Nacional de Diagnóstico y ReferenciaMinistry of HealthManaguaNicaraguaNational Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua.
| |
Collapse
|
154
|
Yang S, Fink D, Hulse A, Pratt RD. Regulatory considerations in development of vaccines to prevent disease caused by Chikungunya virus. Vaccine 2017; 35:4851-4858. [PMID: 28760614 DOI: 10.1016/j.vaccine.2017.07.065] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 06/21/2017] [Accepted: 07/19/2017] [Indexed: 12/01/2022]
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus. Chikungunya disease (CHIK) in humans is characterized by sudden onset of high fever, cutaneous rash, myalgia and debilitating polyarthralgia. Until recently the virus was considered endemic to only Africa and Asia, but since 2004 CHIK has spread to previously non-endemic regions, including Europe and the Americas, thereby emerging as a global health threat. Although a variety of CHIKV vaccine candidates have been tested in animals, and a few have advanced to human clinical trials, no licensed vaccine is currently available for prevention of disease. In this article, we review recent efforts in CHIKV vaccine development and discuss regulatory considerations for CHIKV vaccine licensure under U.S. FDA regulations. Several licensure pathways are available, and the most appropriate licensure pathway for a CHIK vaccine will depend on the type of evidence that can be generated to demonstrate the vaccine's effectiveness. If "traditional approval" following demonstration of direct benefit in adequate and well-controlled clinical disease endpoint studies is not possible, the Accelerated Approval and Animal Rule pathways are potential alternatives. In terms of vaccine safety, the potential for vaccine associated arthralgia and antibody-dependent enhancement of infectivity and disease severity are important issues that should be addressed in both pre-clinical and clinical studies. CHIK vaccine developers are encouraged to communicate with the FDA during all stages of vaccine development.
Collapse
Affiliation(s)
- Sixun Yang
- Division of Vaccines and Related Product Applications, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States.
| | - Doran Fink
- Division of Vaccines and Related Product Applications, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States
| | - Andrea Hulse
- Division of Vaccines and Related Product Applications, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States
| | - R Douglas Pratt
- Division of Vaccines and Related Product Applications, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States
| |
Collapse
|
155
|
Fox JM, Diamond MS. Immune-Mediated Protection and Pathogenesis of Chikungunya Virus. THE JOURNAL OF IMMUNOLOGY 2017; 197:4210-4218. [PMID: 27864552 DOI: 10.4049/jimmunol.1601426] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 09/13/2016] [Indexed: 01/16/2023]
Abstract
Chikungunya virus (CHIKV) is a re-emerging alphavirus that causes debilitating acute and chronic arthritis. Infection by CHIKV induces a robust immune response that is characterized by production of type I IFNs, recruitment of innate and adaptive immune cells, and development of neutralizing Abs. Despite this response, chronic arthritis can develop in some individuals, which may be due to a failure to eliminate viral RNA and Ag and/or persistent immune responses that cause chronic joint inflammation. In this review, based primarily on advances from recent studies in mice, we discuss the innate and adaptive immune factors that control CHIKV dissemination and clearance or contribute to pathogenesis.
Collapse
Affiliation(s)
- Julie M Fox
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110; .,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110.,Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110; and.,Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
156
|
Cirimotich CM, Vela EM, Garver J, Barnewall RE, Miller BD, Meister GT, Rogers JV. Chikungunya virus infection in Cynomolgus macaques following Intradermal and aerosol exposure. Virol J 2017; 14:135. [PMID: 28728590 PMCID: PMC5520379 DOI: 10.1186/s12985-017-0804-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/14/2017] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Chikungunya virus (CHIKV) is transmitted via mosquito bite and potentially by aerosol, causing chikungunya fever and arthritic disease in humans. There are currently no licensed vaccines or antiviral therapeutics to protect against CHIKV infection in humans. Animal models recapitulating human disease, especially for transmission by aerosol, are needed for licensure of such medical countermeasures. METHODS Cynomolgus macaques (CMs) were challenged by intradermal (ID) inoculation or exposure to an aerosol containing CHIKV Ross strain at different target infectious doses (103-107 plaque forming units (PFU)). The clinical and virologic courses of disease were monitored up to 14 days post-exposure. RESULTS ID infection of CMs led to overt clinical disease, detectable viremia, and increased blood markers of liver damage. Animals challenged by aerosol exhibited viremia and increased liver damage biomarkers with minimal observed clinical disease. All animals survived CHIKV challenge. CONCLUSIONS We have described CHIKV infection in CMs following ID inoculation and, for the first time, infection by aerosol. Based on limited reported cases in the published literature, the aerosol model recapitulates the virologic findings of human infection via this route. The results of this study provide additional evidence for the potential use of CMs as a model for evaluating medical countermeasures against CHIKV.
Collapse
Affiliation(s)
| | - Eric M Vela
- Battelle, West Jefferson, OH, 43162, USA.,Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, 97239, USA
| | | | | | | | | | | |
Collapse
|
157
|
Eldi P, Cooper TH, Liu L, Prow NA, Diener KR, Howley PM, Suhrbier A, Hayball JD. Production of a Chikungunya Vaccine Using a CHO Cell and Attenuated Viral-Based Platform Technology. Mol Ther 2017; 25:2332-2344. [PMID: 28720468 PMCID: PMC5628773 DOI: 10.1016/j.ymthe.2017.06.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/03/2017] [Accepted: 06/18/2017] [Indexed: 02/06/2023] Open
Abstract
Vaccinia-based systems have been extensively explored for the development of recombinant vaccines. Herein we describe an innovative vaccinia virus (VACV)-derived vaccine platform technology termed Sementis Copenhagen Vector (SCV), which was rendered multiplication-defective by targeted deletion of the essential viral assembly gene D13L. A SCV cell substrate line was developed for SCV vaccine production by engineering CHO cells to express D13 and the VACV host-range factor CP77, because CHO cells are routinely used for manufacture of biologics. To illustrate the utility of the platform technology, a SCV vaccine against chikungunya virus (SCV-CHIK) was developed and shown to be multiplication-defective in a range of human cell lines and in immunocompromised mice. A single vaccination of mice with SCV-CHIK induced antibody responses specific for chikungunya virus (CHIKV) that were similar to those raised following vaccination with a replication-competent VACV-CHIK and able to neutralize CHIKV. Vaccination also provided protection against CHIKV challenge, preventing both viremia and arthritis. Moreover, SCV retained capacity as an effective mouse smallpox vaccine. In summary, SCV represents a new and safe vaccine platform technology that can be manufactured in modified CHO cells, with pre-clinical evaluation illustrating utility for CHIKV vaccine design and construction.
Collapse
Affiliation(s)
- Preethi Eldi
- Experimental Therapeutics Laboratory, Hanson Institute and Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Tamara H Cooper
- Experimental Therapeutics Laboratory, Hanson Institute and Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Liang Liu
- Experimental Therapeutics Laboratory, Hanson Institute and Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Natalie A Prow
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Kerrilyn R Diener
- Experimental Therapeutics Laboratory, Hanson Institute and Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia; Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Paul M Howley
- Experimental Therapeutics Laboratory, Hanson Institute and Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia; Sementis Ltd., Melbourne, VIC 3000, Australia.
| | - Andreas Suhrbier
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - John D Hayball
- Experimental Therapeutics Laboratory, Hanson Institute and Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia; Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia.
| |
Collapse
|
158
|
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus in the family Togaviridae that causes outbreaks of debilitating acute and chronic arthralgia in humans. Although historically associated with localized outbreaks in Africa and Asia, recent epidemics in the Indian Ocean region and the Americas have led to the recognition that CHIKV is capable of moving into previously unaffected areas and causing significant levels of human suffering. The severity of CHIKV rheumatic disease, which can severely impact life quality of infected individuals for weeks, months, or even years, combined with the explosive nature of CHIKV outbreaks and its demonstrated ability to quickly spread into new regions, has led to renewed interest in developing strategies for the prevention or treatment of CHIKV-induced disease. Therefore, this chapter briefly discusses the biology of CHIKV and the factors contributing to CHIKV dissemination, while also discussing the pathogenesis of CHIKV-induced disease and summarizing the status of efforts to develop safe and effective therapies and vaccines against CHIKV and related viruses.
Collapse
|
159
|
Balmaseda Á, Saborío Galo S, González K, Téllez Y, García N, Pérez L, Gresh L, Harris E. Development of in-house serological methods for diagnosis and surveillance of chikungunya. Rev Panam Salud Publica 2017. [PMID: 28902269 PMCID: PMC5638038 DOI: 10.26633/rpsp.2017.56] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Objective. To develop and evaluate serological methods for chikungunya diagnosis and research in Nicaragua. Methods. Two IgM ELISA capture systems (MAC-ELISA) for diagnosis of acute chikungunya virus (CHIKV) infections, and two Inhibition ELISA Methods (IEM) to measure total antibodies against CHIKV were developed using monoclonal antibodies (mAbs) and hyperimmune serum at the National Virology Laboratory of Nicaragua in 2014–2015. The sensitivity, specificity, predictive values, and agreement of the MAC-ELISAs were obtained by comparing the results of 198 samples (116 positive; 82 negative) with the Centers for Disease Control and Prevention’s IgM ELISA (Atlanta, Georgia, United States; CDC-MAC-ELISA). For clinical evaluation of the four serological techniques, 260 paired acute and convalescent phase serum samples of suspected chikungunya cases were used. Results. All four assays were standardized by determining the optimal concentrations of the different reagents. Processing times were substantially reduced compared to the CDC-MAC-ELISA. For the MAC-ELISA systems, a sensitivity of 96.6% and 97.4%, and a specificity of 98.8% and 91.5% were obtained using mAb and hyperimmune serum, respectively, compared with the CDC method. Clinical evaluation of the four serological techniques versus the CDC real-time RT-PCR assay resulted in a sensitivity of 95.7% and a specificity of 88.8%–95.9%. Conclusion. Two MAC-ELISA and two IEM systems were standardized, demonstrating very good quality for chikungunya diagnosis and research demands. This will achieve more efficient epidemiological surveillance in Nicaragua, the first country in Central America to produce its own reagents for serological diagnosis of CHIKV. The methods evaluated here can be applied in other countries and will contribute to sustainable diagnostic systems to combat the disease.
Collapse
Affiliation(s)
- Ángel Balmaseda
- National Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Saira Saborío Galo
- National Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Karla González
- National Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Yolanda Téllez
- National Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Nadezna García
- National Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Leonel Pérez
- National Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Lionel Gresh
- Sustainable Sciences Institute, Managua, Nicaragua
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, California, United States of America
| |
Collapse
|
160
|
Abstract
Chikungunya virus (CHIKV) is an arthropod-borne alphavirus that causes acute and chronic arthritis. The virus reemerged in the Indian Ocean islands in 2005-2006 and is responsible for outbreaks in the Caribbean islands and the Americas since late 2013. Despite the wealth of research over the past 10 years, there are no commercially available antiviral drugs or vaccines. Treatment usually involves analgesics, anti-inflammatory drugs, and supportive care. Most studies have been focused on understanding the pathogenesis of CHIKV infection through clinical observation and with animal models. In this review, the clinical manifestations of CHIKV that define the disease and the use of relevant animal models, from mice to nonhuman primates, are discussed. Understanding key cellular factors in CHIKV infection and the interplay with the immune system will aid in the development of preventive and therapeutic approaches to combat this painful viral disease in humans.
Collapse
Affiliation(s)
- Lisa F P Ng
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore 138648; .,Institute of Infection and Global Health, University of Liverpool, Liverpool L69 3BX, United Kingdom
| |
Collapse
|
161
|
Therapeutic administration of a recombinant human monoclonal antibody reduces the severity of chikungunya virus disease in rhesus macaques. PLoS Negl Trop Dis 2017. [PMID: 28628616 PMCID: PMC5491320 DOI: 10.1371/journal.pntd.0005637] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne virus that causes a febrile syndrome in humans associated with acute and chronic debilitating joint and muscle pain. Currently no licensed vaccines or therapeutics are available to prevent or treat CHIKV infections. We recently isolated a panel of potently neutralizing human monoclonal antibodies (mAbs), one (4N12) of which exhibited prophylactic and post-exposure therapeutic activity against CHIKV in immunocompromised mice. Here, we describe the development of an engineered CHIKV mAb, designated SVIR001, that has similar antigen binding and neutralization profiles to its parent, 4N12. Because therapeutic administration of SVIR001 in immunocompetent mice significantly reduced viral load in joint tissues, we evaluated its efficacy in a rhesus macaque model of CHIKV infection. Rhesus macaques that were treated after infection with SVIR001 showed rapid elimination of viremia and less severe joint infiltration and disease compared to animals treated with SVIR002, an isotype control mAb. SVIR001 reduced viral burden at the site of infection and at distant sites and also diminished the numbers of activated innate immune cells and levels of pro-inflammatory cytokines and chemokines. SVIR001 therapy; however, did not substantively reduce the induction of CHIKV-specific B or T cell responses. Collectively, these results show promising therapeutic activity of a human anti-CHIKV mAb in rhesus macaques and provide proof-of-principle for its possible use in humans to treat active CHIKV infections. Chikungunya virus (CHIKV) causes fever, rash, and acute and chronic arthralgia. Currently there are no approved therapies to treat or vaccines to prevent CHIKV infection in humans. In this study, we engineered SVIR001, a recombinant fully human monoclonal antibody (mAb) that eliminated viremia, reduced viral load at the site of infection, and diminished spread to distant target tissues in rhesus macaques when administered after infection. SVIR001 treatment reduced joint inflammation and disease without impairing the induction of the adaptive immune response. These results demonstrate the efficacy of mAb therapy to reduce the severity of CHIKV disease.
Collapse
|
162
|
DeZure AD, Berkowitz NM, Graham BS, Ledgerwood JE. Whole-Inactivated and Virus-Like Particle Vaccine Strategies for Chikungunya Virus. J Infect Dis 2017; 214:S497-S499. [PMID: 27920180 DOI: 10.1093/infdis/jiw352] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Chikungunya virus (CHIKV) is a global public health threat, having been identified in >60 countries in Asia, Africa, Europe, and the Americas. There is no cure for or licensed vaccine against CHIKV infection. Initial attempts at CHIKV vaccine development began in the early 1960s. Whole-inactivated and virus-like particle (VLP) vaccines are 2 of the current approaches being evaluated. Success of these approaches is dependent on a safe, well-tolerated vaccine that is immunogenic and deployable in regard to manufacturing, stability, and delivery characteristics.
Collapse
Affiliation(s)
- Adam D DeZure
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Nina M Berkowitz
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Julie E Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
163
|
Abstract
Chikungunya is caused by an alphavirus that is transmitted to humans via the Aedes species mosquito. Chikungunya is endemic to tropical Africa and South and Southeast Asia, but over the past decade, the geographic distribution of the virus has been expanding rapidly. The disease is characterized by fever and severe polyarthritis, and although symptoms typically resolve within 7 to 10 days, some patients experience persistent arthritis and arthralgias for months to years.In December 2013, the first local transmission of chikungunya virus in the Americas was identified in the Caribbean Island of Saint Martin. Since then, the number of afflicted individuals has spread throughout the Caribbean and Central America, as well as into South America. The United States reported 2788 chikungunya virus disease cases among travelers returning from affected areas in 2014. In addition, 11 locally acquired cases were reported in Florida. Further spread and establishment of the disease in the Americas are likely considering the high levels of viremia in infected individuals, widespread distribution of effective vectors, lack of immunity among people living in the Americas, and the popularity of international travel.Considering the prominent rheumatic manifestations of chikungunya, rheumatologists are likely to encounter patients with the disease in their practice. We recommend that rheumatologists consider chikungunya in their differential diagnosis when evaluating patients presenting with fever and joint pain following travel to a chikungunya endemic area. Early diagnosis would ensure timely management and reduction of polypharmacy and its associated complications. In this article, we briefly describe the epidemiology of chikungunya, the clinical features, laboratory testing, prevention, and treatment of disease.
Collapse
|
164
|
Haese NN, Broeckel RM, Hawman DW, Heise MT, Morrison TE, Streblow DN. Animal Models of Chikungunya Virus Infection and Disease. J Infect Dis 2017; 214:S482-S487. [PMID: 27920178 DOI: 10.1093/infdis/jiw284] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Chikungunya virus (CHIKV) is a reemerging alphavirus that causes acute febrile illness and severe joint pain in humans. Although acute symptoms often resolve within a few days, chronic joint and muscle pain can be long lasting. In the last decade, CHIKV has caused widespread outbreaks of unprecedented scale in the Americas, Asia, and the Indian Ocean island regions. Despite these outbreaks and the continued expansion of CHIKV into new areas, mechanisms of chikungunya pathogenesis and disease are not well understood. Experimental animal models are indispensable to the field of CHIKV research. The most commonly used experimental animal models of CHIKV infection are mice and nonhuman primates; each model has its advantages for studying different aspects of CHIKV disease. This review will provide an overview of animal models used to study CHIKV infection and disease and major advances in our understanding of chikungunya obtained from studies performed in these models.
Collapse
Affiliation(s)
- Nicole N Haese
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton
| | - Rebecca M Broeckel
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton
| | - David W Hawman
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora
| | - Mark T Heise
- Departments of Genetics, Microbiology, and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
| | - Thomas E Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton
| |
Collapse
|
165
|
Clayton AM. Monoclonal Antibodies as Prophylactic and Therapeutic Agents Against Chikungunya Virus. J Infect Dis 2017; 214:S506-S509. [PMID: 27920182 DOI: 10.1093/infdis/jiw324] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that is responsible for considerable epidemics worldwide and recently emerged in the Americas in 2013. CHIKV may cause long-lasting arthralgia after acute infection. With currently no licensed vaccines or antivirals, the design of effective therapies to prevent or treat CHIKV infection is of utmost importance and will be facilitated by increased understanding of the dynamics of chikungunya. In this article, monoclonal antibodies against CHIKV as viable prophylactic and therapeutic agents will be discussed.
Collapse
Affiliation(s)
- April M Clayton
- Office of Global Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
166
|
Gayen M, Gupta P, Morazzani EM, Gaidamakova EK, Knollmann-Ritschel B, Daly MJ, Glass PJ, Maheshwari RK. Deinococcus Mn 2+-peptide complex: A novel approach to alphavirus vaccine development. Vaccine 2017; 35:3672-3681. [PMID: 28576570 DOI: 10.1016/j.vaccine.2017.05.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 04/17/2017] [Accepted: 05/04/2017] [Indexed: 10/19/2022]
Abstract
Over the last ten years, Chikungunya virus (CHIKV), an Old World alphavirus has caused numerous outbreaks in Asian and European countries and the Americas, making it an emerging pathogen of great global health importance. Venezuelan equine encephalitis virus (VEEV), a New World alphavirus, on the other hand, has been developed as a bioweapon in the past due to its ease of preparation, aerosol dispersion and high lethality in aerosolized form. Currently, there are no FDA approved vaccines against these viruses. In this study, we used a novel approach to develop inactivated vaccines for VEEV and CHIKV by applying gamma-radiation together with a synthetic Mn-decapeptide-phosphate complex (MnDpPi), based on manganous-peptide-orthophosphate antioxidants accumulated in the extremely radiation-resistant bacterium Deinococcus radiodurans. Classical gamma-irradiated vaccine development approaches are limited by immunogenicity-loss due to oxidative damage to the surface proteins at the high doses of radiation required for complete virus-inactivation. However, addition of MnDpPi during irradiation process selectively protects proteins, but not the nucleic acids, from the radiation-induced oxidative damage, as required for safe and efficacious vaccine development. Previously, this approach was used to develop a bacterial vaccine. In the present study, we show that this approach can successfully be applied to protecting mice against viral infections. Irradiation of VEEV and CHIKV in the presence of MnDpPi resulted in substantial epitope preservation even at supra-lethal doses of gamma-rays (50,000Gy). Irradiated viruses were found to be completely inactivated and safe in vivo (neonatal mice). Upon immunization, VEEV inactivated in the presence of MnDpPi resulted in drastically improved protective efficacy. Thus, the MnDpPi-based gamma-inactivation approach described here can readily be applied to developing vaccines against any pathogen of interest in a fast and cost-effective manner.
Collapse
Affiliation(s)
- Manoshi Gayen
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Henry M. Jackson Foundation, Bethesda, MD 20817, USA
| | - Paridhi Gupta
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Henry M. Jackson Foundation, Bethesda, MD 20817, USA.
| | - Elaine M Morazzani
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Elena K Gaidamakova
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Henry M. Jackson Foundation, Bethesda, MD 20817, USA
| | | | - Michael J Daly
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| | - Pamela J Glass
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Radha K Maheshwari
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
167
|
Roques P, Ljungberg K, Kümmerer BM, Gosse L, Dereuddre-Bosquet N, Tchitchek N, Hallengärd D, García-Arriaza J, Meinke A, Esteban M, Merits A, Le Grand R, Liljeström P. Attenuated and vectored vaccines protect nonhuman primates against Chikungunya virus. JCI Insight 2017; 2:e83527. [PMID: 28352649 DOI: 10.1172/jci.insight.83527] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Chikungunya virus (CHIKV) is rapidly spreading across the globe, and millions are infected. Morbidity due to this virus is a serious threat to public health, but at present, there is no vaccine against this debilitating disease. We have recently developed a number of vaccine candidates, and here we have evaluated 3 of them in a nonhuman primate model. A single immunization with an attenuated strain of CHIKV (Δ5nsP3), a homologous prime-boost immunization with a DNA-launched RNA replicon encoding CHIKV envelope proteins (DREP-E), and a DREP-E prime followed by a recombinant modified vaccinia virus Ankara encoding CHIKV capsid and envelope (MVA-CE) boost all induced protection against WT CHIKV infection. The attenuated Δ5nsP3 virus proved to be safe and did not show any clinical signs typically associated with WT CHIKV infections such as fever, skin rash, lymphopenia, or joint swelling. These vaccines are based on an East/Central/South African strain of Indian Ocean lineage, but they also generated neutralizing antibodies against an isolate of the Asian genotype that now is rapidly spreading across the Americas. These results form the basis for clinical development of an efficacious CHIKV vaccine that generates both humoral and cellular immunity with long-term immunological memory.
Collapse
Affiliation(s)
- Pierre Roques
- Université Paris Sud, UMR 1184, Orsay, France.,CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT center.,Inserm, U1184, Center for immunology of viral infections and autoimmune diseases, Fontenay aux Roses, France
| | - Karl Ljungberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Beate M Kümmerer
- Institute of Virology, University of Bonn Medical Centre, Bonn, Germany
| | - Leslie Gosse
- Université Paris Sud, UMR 1184, Orsay, France.,CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT center.,Inserm, U1184, Center for immunology of viral infections and autoimmune diseases, Fontenay aux Roses, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris Sud, UMR 1184, Orsay, France.,CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT center.,Inserm, U1184, Center for immunology of viral infections and autoimmune diseases, Fontenay aux Roses, France
| | - Nicolas Tchitchek
- Université Paris Sud, UMR 1184, Orsay, France.,CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT center.,Inserm, U1184, Center for immunology of viral infections and autoimmune diseases, Fontenay aux Roses, France
| | - David Hallengärd
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | | | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Andres Merits
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Roger Le Grand
- Université Paris Sud, UMR 1184, Orsay, France.,CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT center.,Inserm, U1184, Center for immunology of viral infections and autoimmune diseases, Fontenay aux Roses, France
| | - Peter Liljeström
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
168
|
Silva LA, Dermody TS. Chikungunya virus: epidemiology, replication, disease mechanisms, and prospective intervention strategies. J Clin Invest 2017; 127:737-749. [PMID: 28248203 PMCID: PMC5330729 DOI: 10.1172/jci84417] [Citation(s) in RCA: 251] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Chikungunya virus (CHIKV), a reemerging arbovirus, causes a crippling musculoskeletal inflammatory disease in humans characterized by fever, polyarthralgia, myalgia, rash, and headache. CHIKV is transmitted by Aedes species of mosquitoes and is capable of an epidemic, urban transmission cycle with high rates of infection. Since 2004, CHIKV has spread to new areas, causing disease on a global scale, and the potential for CHIKV epidemics remains high. Although CHIKV has caused millions of cases of disease and significant economic burden in affected areas, no licensed vaccines or antiviral therapies are available. In this Review, we describe CHIKV epidemiology, replication cycle, pathogenesis and host immune responses, and prospects for effective vaccines and highlight important questions for future research.
Collapse
|
169
|
The neutralizing role of IgM during early Chikungunya virus infection. PLoS One 2017; 12:e0171989. [PMID: 28182795 PMCID: PMC5300252 DOI: 10.1371/journal.pone.0171989] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/30/2017] [Indexed: 12/13/2022] Open
Abstract
The antibody isotype IgM appears earlier than IgG, within days of onset of symptoms, and is important during the early stages of the adaptive immune response. Little is known about the functional role of IgM during infection with chikungunya virus (CHIKV), a recently reemerging arbovirus that has caused large global outbreaks. In this study, we studied antibody responses in 102 serum samples collected during CHIKV outbreaks in Malaysia. We described the neutralizing role of IgM at different times post-infection and examined the independent contributions of IgM and IgG towards the neutralizing capacity of human immune sera during the early phase of infection, including the differences in targets of neutralizing epitopes. Neutralizing IgM starts to appear as early as day 4 of symptoms, and their appearance from day 6 is associated with a reduction in viremia. IgM acts in a complementary manner with the early IgG, but plays the main neutralizing role up to a point between days 4 and 10 which varies between individuals. After this point, total neutralizing capacity is attributable almost entirely to the robust neutralizing IgG response. IgM preferentially binds and targets epitopes on the CHIKV surface E1-E2 glycoproteins, rather than individual E1 or E2. These findings provide insight into the early antibody responses to CHIKV, and have implications for design of diagnostic serological assays.
Collapse
|
170
|
Miner JJ, Cook LE, Hong JP, Smith AM, Richner JM, Shimak RM, Young AR, Monte K, Poddar S, Crowe JE, Lenschow DJ, Diamond MS. Therapy with CTLA4-Ig and an antiviral monoclonal antibody controls chikungunya virus arthritis. Sci Transl Med 2017; 9:eaah3438. [PMID: 28148840 PMCID: PMC5448557 DOI: 10.1126/scitranslmed.aah3438] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/21/2016] [Accepted: 12/02/2016] [Indexed: 12/12/2022]
Abstract
In 2013, chikungunya virus (CHIKV) transmission was documented in the Western Hemisphere, and the virus has since spread throughout the Americas with more than 1.8 million people infected in more than 40 countries. CHIKV targets the joints, resulting in symmetric polyarthritis that clinically mimics rheumatoid arthritis and can endure for months to years. At present, no approved treatment is effective in preventing or controlling CHIKV infection or disease. We treated mice with eight different disease-modifying antirheumatic drugs and identified CLTA4-Ig (abatacept) and tofacitinib as candidate therapies based on their ability to decrease acute joint swelling. CTLA4-Ig reduced T cell accumulation in the joints of infected animals without affecting viral infection. Whereas monotherapy with CTLA4-Ig or a neutralizing anti-CHIKV human monoclonal antibody provided partial clinical improvement, therapy with both abolished swelling and markedly reduced levels of chemokines, proinflammatory cytokines, and infiltrating leukocytes. Thus, combination CTLA4-Ig and antiviral antibody therapy controls acute CHIKV infection and arthritis and may be a candidate for testing in humans.
Collapse
Affiliation(s)
- Jonathan J Miner
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lindsey E Cook
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jun P Hong
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Amber M Smith
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Justin M Richner
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Raeann M Shimak
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alissa R Young
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kristen Monte
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Subhajit Poddar
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James E Crowe
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Deborah J Lenschow
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
171
|
Sapparapu G, Fernandez E, Kose N, Bin Cao, Fox JM, Bombardi RG, Zhao H, Nelson CA, Bryan AL, Barnes T, Davidson E, Mysorekar IU, Fremont DH, Doranz BJ, Diamond MS, Crowe JE. Neutralizing human antibodies prevent Zika virus replication and fetal disease in mice. Nature 2016; 540:443-447. [PMID: 27819683 PMCID: PMC5583716 DOI: 10.1038/nature20564] [Citation(s) in RCA: 308] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/27/2016] [Indexed: 02/07/2023]
Abstract
Zika virus (ZIKV) is an emerging mosquito-transmitted flavivirus that can cause severe disease, including congenital birth defects during pregnancy. To develop candidate therapeutic agents against ZIKV, we isolated a panel of human monoclonal antibodies from subjects that were previously infected with ZIKV. We show that a subset of antibodies recognize diverse epitopes on the envelope (E) protein and exhibit potent neutralizing activity. One of the most inhibitory antibodies, ZIKV-117, broadly neutralized infection of ZIKV strains corresponding to African and Asian-American lineages. Epitope mapping studies revealed that ZIKV-117 recognized a unique quaternary epitope on the E protein dimer-dimer interface. We evaluated the therapeutic efficacy of ZIKV-117 in pregnant and non-pregnant mice. Monoclonal antibody treatment markedly reduced tissue pathology, placental and fetal infection, and mortality in mice. Thus, neutralizing human antibodies can protect against maternal-fetal transmission, infection and disease, and reveal important determinants for structure-based rational vaccine design efforts.
Collapse
MESH Headings
- Africa
- Americas
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Viral/chemistry
- Antibodies, Viral/immunology
- Antibodies, Viral/therapeutic use
- Antibody Specificity
- Asia
- B-Lymphocytes/immunology
- Disease Models, Animal
- Epitope Mapping
- Female
- Fetal Diseases/immunology
- Fetal Diseases/prevention & control
- Fetal Diseases/virology
- Fetus/immunology
- Fetus/virology
- Humans
- Infectious Disease Transmission, Vertical/prevention & control
- Male
- Mice
- Models, Molecular
- Placenta/immunology
- Placenta/virology
- Pregnancy
- Protein Multimerization
- Survival Rate
- Viral Proteins/chemistry
- Viral Proteins/immunology
- Viral Vaccines/chemistry
- Viral Vaccines/immunology
- Virus Replication/immunology
- Zika Virus/growth & development
- Zika Virus/immunology
- Zika Virus Infection/immunology
- Zika Virus Infection/pathology
- Zika Virus Infection/virology
Collapse
Affiliation(s)
- Gopal Sapparapu
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Estefania Fernandez
- Department of Pathology &Immunology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Nurgun Kose
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bin Cao
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Julie M Fox
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
| | - Robin G Bombardi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Haiyan Zhao
- Department of Pathology &Immunology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Christopher A Nelson
- Department of Pathology &Immunology, Washington University School of Medicine, St Louis, Missouri, USA
| | | | | | | | - Indira U Mysorekar
- Department of Pathology &Immunology, Washington University School of Medicine, St Louis, Missouri, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Daved H Fremont
- Department of Pathology &Immunology, Washington University School of Medicine, St Louis, Missouri, USA
| | | | - Michael S Diamond
- Department of Pathology &Immunology, Washington University School of Medicine, St Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, USA
- Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, Missouri, USA
| | - James E Crowe
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
172
|
Abstract
Bispecific antibody engineering, in which binding specificities toward 2 distinct epitopes are combined into a single molecule, can greatly enhance immunotherapeutic properties of monoclonal antibodies. While the bispecific antibody approach has been applied widely to targets for indications such as cancer and inflammation, the development of such agents for viral immunotherapy is only now emerging. Here, we review recent advances in the development of bispecific antibodies for viral immunotherapy, highlighting promising in vitro and in vivo results.
Collapse
Affiliation(s)
- Elisabeth K Nyakatura
- a Department of Biochemistry , Albert Einstein College of Medicine , Bronx , NY , USA
| | - Alexandra Y Soare
- a Department of Biochemistry , Albert Einstein College of Medicine , Bronx , NY , USA
| | - Jonathan R Lai
- a Department of Biochemistry , Albert Einstein College of Medicine , Bronx , NY , USA
| |
Collapse
|
173
|
Dysregulated TGF-β Production Underlies the Age-Related Vulnerability to Chikungunya Virus. PLoS Pathog 2016; 12:e1005891. [PMID: 27736984 PMCID: PMC5063327 DOI: 10.1371/journal.ppat.1005891] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 08/23/2016] [Indexed: 02/06/2023] Open
Abstract
Chikungunya virus (CHIKV) is a re-emerging global pathogen with pandemic potential, which causes fever, rash and debilitating arthralgia. Older adults over 65 years are particularly susceptible to severe and chronic CHIKV disease (CHIKVD), accounting for >90% of all CHIKV-related deaths. There are currently no approved vaccines or antiviral treatments available to limit chronic CHIKVD. Here we show that in old mice excessive, dysregulated TGFβ production during acute infection leads to a reduced immune response and subsequent chronic disease. Humans suffering from CHIKV infection also exhibited high TGFβ levels and a pronounced age-related defect in neutralizing anti-CHIKV antibody production. In vivo reduction of TGFβ levels minimized acute joint swelling, restored neutralizing antibody production and diminished chronic joint pathology in old mice. This study identifies increased and dysregulated TGFβ secretion as one key mechanism contributing to the age-related loss of protective anti-CHIKV-immunity leading to chronic CHIKVD.
Collapse
|
174
|
Kuo SC, Wang YM, Ho YJ, Chang TY, Lai ZZ, Tsui PY, Wu TY, Lin CC. Suramin treatment reduces chikungunya pathogenesis in mice. Antiviral Res 2016; 134:89-96. [DOI: 10.1016/j.antiviral.2016.07.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 07/19/2016] [Accepted: 07/19/2016] [Indexed: 12/29/2022]
|
175
|
Goo L, Dowd KA, Lin TY, Mascola JR, Graham BS, Ledgerwood JE, Pierson TC. A Virus-Like Particle Vaccine Elicits Broad Neutralizing Antibody Responses in Humans to All Chikungunya Virus Genotypes. J Infect Dis 2016; 214:1487-1491. [PMID: 27655868 DOI: 10.1093/infdis/jiw431] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/07/2016] [Indexed: 12/19/2022] Open
Abstract
Chikungunya virus (CHIKV) is an alphavirus that has emerged as a global health burden. There are 3 CHIKV genotypes: Asian, West African, and Eastern/Central/South African. No licensed CHIKV vaccine is available, and whether the antibody response elicited by one genotype can neutralize heterologous genotypes is unclear. We assessed neutralizing antibody (NAb) responses of volunteers in a phase 1 study of a CHIKV vaccine against 9 viral strains representing all 3 genotypes. Minimal differences in vaccine-elicited NAb responses were observed among genotypes, suggesting that vaccination with a single CHIKV strain can elicit cross-protective NAbs against all 3 genotypes.
Collapse
Affiliation(s)
- Leslie Goo
- Viral Pathogenesis Section, Laboratory of Viral Diseases
| | | | - Tsai-Yu Lin
- Viral Pathogenesis Section, Laboratory of Viral Diseases
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Julie E Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | | |
Collapse
|
176
|
The Interferon-Stimulated Gene IFITM3 Restricts Infection and Pathogenesis of Arthritogenic and Encephalitic Alphaviruses. J Virol 2016; 90:8780-94. [PMID: 27440901 DOI: 10.1128/jvi.00655-16] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/17/2016] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED Host cells respond to viral infections by producing type I interferon (IFN), which induces the expression of hundreds of interferon-stimulated genes (ISGs). Although ISGs mediate a protective state against many pathogens, the antiviral functions of the majority of these genes have not been identified. IFITM3 is a small transmembrane ISG that restricts a broad range of viruses, including orthomyxoviruses, flaviviruses, filoviruses, and coronaviruses. Here, we show that alphavirus infection is increased in Ifitm3(-/-) and Ifitm locus deletion (Ifitm-del) fibroblasts and, reciprocally, reduced in fibroblasts transcomplemented with Ifitm3. Mechanistic studies showed that Ifitm3 did not affect viral binding or entry but inhibited pH-dependent fusion. In a murine model of chikungunya virus arthritis, Ifitm3(-/-) mice sustained greater joint swelling in the ipsilateral ankle at days 3 and 7 postinfection, and this correlated with higher levels of proinflammatory cytokines and viral burden. Flow cytometric analysis suggested that Ifitm3(-/-) macrophages from the spleen were infected at greater levels than observed in wild-type (WT) mice, results that were supported by experiments with Ifitm3(-/-) bone marrow-derived macrophages. Ifitm3(-/-) mice also were more susceptible than WT mice to lethal alphavirus infection with Venezuelan equine encephalitis virus, and this was associated with greater viral burden in multiple organs. Collectively, our data define an antiviral role for Ifitm3 in restricting infection of multiple alphaviruses. IMPORTANCE The interferon-induced transmembrane protein 3 (IFITM3) inhibits infection of multiple families of viruses in cell culture. Compared to other viruses, much less is known about the antiviral effect of IFITM3 on alphaviruses. In this study, we characterized the antiviral activity of mouse Ifitm3 against arthritogenic and encephalitic alphaviruses using cells and animals with a targeted gene deletion of Ifitm3 as well as deficient cells transcomplemented with Ifitm3. Based on extensive virological analysis, we demonstrate greater levels of alphavirus infection and disease pathogenesis when Ifitm3 expression is absent. Our data establish an inhibitory role for Ifitm3 in controlling infection of alphaviruses.
Collapse
|
177
|
Zhao H, Fernandez E, Dowd KA, Speer SD, Platt DJ, Gorman MJ, Govero J, Nelson CA, Pierson TC, Diamond MS, Fremont DH. Structural Basis of Zika Virus-Specific Antibody Protection. Cell 2016; 166:1016-1027. [PMID: 27475895 PMCID: PMC4983199 DOI: 10.1016/j.cell.2016.07.020] [Citation(s) in RCA: 299] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 07/14/2016] [Accepted: 07/14/2016] [Indexed: 11/18/2022]
Abstract
Zika virus (ZIKV) infection during pregnancy has emerged as a global public health problem because of its ability to cause severe congenital disease. Here, we developed six mouse monoclonal antibodies (mAbs) against ZIKV including four (ZV-48, ZV-54, ZV-64, and ZV-67) that were ZIKV specific and neutralized infection of African, Asian, and American strains to varying degrees. X-ray crystallographic and competition binding analyses of Fab fragments and scFvs defined three spatially distinct epitopes in DIII of the envelope protein corresponding to the lateral ridge (ZV-54 and ZV-67), C-C' loop (ZV-48 and ZV-64), and ABDE sheet (ZV-2) regions. In vivo passive transfer studies revealed protective activity of DIII-lateral ridge specific neutralizing mAbs in a mouse model of ZIKV infection. Our results suggest that DIII is targeted by multiple type-specific antibodies with distinct neutralizing activity, which provides a path for developing prophylactic antibodies for use in pregnancy or designing epitope-specific vaccines against ZIKV.
Collapse
Affiliation(s)
- Haiyan Zhao
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Estefania Fernandez
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Kimberly A Dowd
- Viral Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Scott D Speer
- Viral Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Derek J Platt
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Matthew J Gorman
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jennifer Govero
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Christopher A Nelson
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; The Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Theodore C Pierson
- Viral Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael S Diamond
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; The Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - Daved H Fremont
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO 63110, USA; The Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
178
|
Hawman DW, Fox JM, Ashbrook AW, May NA, Schroeder KMS, Torres RM, Crowe JE, Dermody TS, Diamond MS, Morrison TE. Pathogenic Chikungunya Virus Evades B Cell Responses to Establish Persistence. Cell Rep 2016; 16:1326-1338. [PMID: 27452455 DOI: 10.1016/j.celrep.2016.06.076] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 06/09/2016] [Accepted: 06/17/2016] [Indexed: 10/21/2022] Open
Abstract
Chikungunya virus (CHIKV) and related alphaviruses cause epidemics of acute and chronic musculoskeletal disease. To investigate the mechanisms underlying the failure of immune clearance of CHIKV, we studied mice infected with an attenuated CHIKV strain (181/25) and the pathogenic parental strain (AF15561), which differ by five amino acids. Whereas AF15561 infection of wild-type mice results in viral persistence in joint tissues, 181/25 is cleared. In contrast, 181/25 infection of μMT mice lacking mature B cells results in viral persistence in joint tissues, suggesting that virus-specific antibody is required for clearance of infection. Mapping studies demonstrated that a highly conserved glycine at position 82 in the A domain of the E2 glycoprotein impedes clearance and neutralization of multiple CHIKV strains. Remarkably, murine and human antibodies targeting E2 domain B failed to neutralize pathogenic CHIKV strains efficiently. Our data suggest that pathogenic CHIKV strains evade E2 domain-B-neutralizing antibodies to establish persistence.
Collapse
Affiliation(s)
- David W Hawman
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Julie M Fox
- Department of Medicine, School of Medicine, Washington University, Saint Louis, MO 63110, USA
| | - Alison W Ashbrook
- Department of Pediatrics, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA; Elizabeth B. Lamb Center for Pediatric Research, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Nicholas A May
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Kristin M S Schroeder
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Raul M Torres
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - James E Crowe
- Department of Pediatrics, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Terence S Dermody
- Department of Pediatrics, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA; Elizabeth B. Lamb Center for Pediatric Research, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, School of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Michael S Diamond
- Department of Medicine, School of Medicine, Washington University, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, School of Medicine, Washington University, Saint Louis, MO 63110, USA; Department of Pathology & Immunology, School of Medicine, Washington University, Saint Louis, MO 63110, USA; Center for Human Immunology and Immunotherapy Programs and School of Medicine, Washington University, Saint Louis, MO 63110, USA
| | - Thomas E Morrison
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO 80045, USA.
| |
Collapse
|
179
|
Adouchief S, Smura T, Vapalahti O, Hepojoki J. Mapping of human B-cell epitopes of Sindbis virus. J Gen Virol 2016; 97:2243-2254. [PMID: 27339177 DOI: 10.1099/jgv.0.000531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mosquito-transmitted Sindbis virus (SINV) causes fever, skin lesions and musculoskeletal symptoms if transmitted to man. SINV is the prototype virus of genus Alphavirus, which includes other arthritogenic viruses such as chikungunya virus (CHIKV) and Ross River virus (RRV) that cause large epidemics with a considerable public health burden. Until now the human B-cell epitopes have been studied for CHIKV and RRV, but not for SINV. To identify the B-cell epitopes in SINV-infection, we synthetised a library of linear 18-mer peptides covering the structural polyprotein of SINV, and probed it with SINV IgG-positive and IgG-negative serum pools. By comparing the binding profiles of the pools, we identified 15 peptides that were strongly reactive only with the SINV IgG-positive pools. We then utilized alanine scanning and individual (n=22) patient sera to further narrow the number of common B-cell epitopes to six. These epitopes locate to the capsid, E2, E1 and to a region in PE2 (uncleaved E3-E2), which may only be present in immature virions. By sequence comparison, we observed that one of the capsid protein epitopes shares six identical amino acids with macrophage migration inhibitory factor (MIF) receptor, which is linked to inflammatory diseases and to molecular pathology of alphaviral arthritides. Our results add to the current understanding on SINV disease and raise questions of a potential role of uncleaved PE2 and the MIF receptor (CD74) mimotope in human SINV infection.
Collapse
Affiliation(s)
- Samuel Adouchief
- Department of Virology, Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| | - Teemu Smura
- Department of Virology, Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| | - Olli Vapalahti
- Department of Virology, Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland.,Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland.,Department of Virology and Immunology, Hospital District of Helsinki and Uusimaa (HUSLAB), Helsinki, Finland
| | - Jussi Hepojoki
- Department of Virology, Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| |
Collapse
|
180
|
Kuan G, Ramirez S, Gresh L, Ojeda S, Melendez M, Sanchez N, Collado D, Garcia N, Mercado JC, Gordon A, Balmaseda A, Harris E. Seroprevalence of Anti-Chikungunya Virus Antibodies in Children and Adults in Managua, Nicaragua, After the First Chikungunya Epidemic, 2014-2015. PLoS Negl Trop Dis 2016; 10:e0004773. [PMID: 27322692 PMCID: PMC4913910 DOI: 10.1371/journal.pntd.0004773] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 05/20/2016] [Indexed: 01/25/2023] Open
Abstract
Chikungunya is a viral disease transmitted by Aedes aegypti and Ae. albopictus mosquitoes. In late 2013, chikungunya virus (CHIKV) was introduced into the Caribbean island of St. Martin. Since then, approximately 2 million chikungunya cases have been reported by the Pan American Health Organization, and most countries in the Americas report autochthonous transmission of CHIKV. In Nicaragua, the first imported case was described in July 2014 and the first autochthonous case in September 2014. Here, we conducted two studies to analyze the seroprevalence of anti-CHIKV antibodies after the first chikungunya epidemic in a community-based cohort study (ages 2-14 years) and in a cross-sectional survey of persons aged ≥15 years in the same area of Managua, Nicaragua. Routine annual serum samples collected from 3,362 cohort participants in March/April 2014 and 2015, and 848 age-stratified samples collected from persons ≥15 years old at the end of May-beginning of June 2015 were used to estimate the seroprevalence of anti-CHIKV antibodies after the first epidemic (October 2014 to February 2015 in the study population). Using an Inhibition ELISA assay that measures total anti-CHIKV antibodies, the seroprevalence was significantly higher in those aged ≥15 (13.1% (95%CI: 10.9, 15.5)) than in the pediatric population (6.1% (95%CI: 5.3, 6.9)). The proportion of inapparent infections was 58.3% (95%CI: 51.5, 65.1) in children and 64.9% (95%CI: 55.2, 73.7) in the ≥15 study population. We identified age, water availability, household size, and socioeconomic status as factors associated with the presence of anti-CHIKV antibodies. Overall, this is the first report of CHIKV seropositivity in continental Latin America and provides useful information for public health authorities in the region.
Collapse
Affiliation(s)
- Guillermina Kuan
- Centro de Salud Sócrates Flores Vivas, Ministry of Health, Managua, Nicaragua
| | - Stephania Ramirez
- Sustainable Sciences Institute, Managua, Nicaragua
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, United States of America
| | - Lionel Gresh
- Sustainable Sciences Institute, Managua, Nicaragua
| | - Sergio Ojeda
- Sustainable Sciences Institute, Managua, Nicaragua
| | | | - Nery Sanchez
- Sustainable Sciences Institute, Managua, Nicaragua
| | - Damaris Collado
- National Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Nadezna Garcia
- National Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Juan Carlos Mercado
- National Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Aubree Gordon
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, United States of America
| | - Angel Balmaseda
- National Virology Laboratory, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, United States of America
- * E-mail:
| |
Collapse
|
181
|
A CRISPR screen defines a signal peptide processing pathway required by flaviviruses. Nature 2016; 535:164-8. [PMID: 27383988 PMCID: PMC4945490 DOI: 10.1038/nature18625] [Citation(s) in RCA: 289] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 06/06/2016] [Indexed: 12/17/2022]
Abstract
Flaviviruses infect hundreds of millions of people annually, and no antiviral therapy is available. We performed a genome-wide CRISPR/Cas9-based screen to identify host genes that, when edited, resulted in reduced flavivirus infection. Here, we validated nine human genes required for flavivirus infectivity, and these were associated with endoplasmic reticulum functions including translocation, protein degradation, and N-linked glycosylation. In particular, a subset of endoplasmic reticulum-associated signal peptidase complex (SPCS) proteins was necessary for proper cleavage of the flavivirus structural proteins (prM and E) and secretion of viral particles. Loss of SPCS1 expression resulted in markedly reduced yield of all Flaviviridae family members tested (West Nile, Dengue, Zika, yellow fever, Japanese encephalitis, and hepatitis C viruses), but had little impact on alphavirus, bunyavirus, or rhabdovirus infection or the surface expression or secretion of diverse host proteins. We found that SPCS1 dependence could be bypassed by replacing the native prM protein leader sequences with a class I major histocompatibility complex (MHC) antigen leader sequence. Thus, SPCS1, either directly or indirectly via its interactions with unknown host proteins, preferentially promotes the processing of specific protein cargo, and Flaviviridae have a unique dependence on this signal peptide processing pathway. SPCS1 and other signal processing pathway members could represent pharmacological targets for inhibiting infection by the expanding number of flaviviruses of medical concern.
Collapse
|
182
|
Plasmodium falciparum Histidine-Rich Protein II Compromises Brain Endothelial Barriers and May Promote Cerebral Malaria Pathogenesis. mBio 2016; 7:mBio.00617-16. [PMID: 27273825 PMCID: PMC4959673 DOI: 10.1128/mbio.00617-16] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cerebral malaria (CM) is a disease of the vascular endothelium caused by Plasmodium falciparum. It is characterized by parasite sequestration, inflammatory cytokine production, and vascular leakage. A distinguishing feature of P. falciparum infection is parasite production and secretion of histidine-rich protein II (HRPII). Plasma HRPII is a diagnostic and prognostic marker for falciparum malaria. We demonstrate that disruption of a human cerebral microvascular endothelial barrier by P. falciparum-infected erythrocytes depends on expression of HRPII. Purified recombinant or native HRPII can recapitulate these effects. HRPII action occurs via activation of the inflammasome, resulting in decreased integrity of tight junctions and increased endothelial permeability. We propose that HRPII is a virulence factor that may contribute to cerebral malaria by compromising endothelial barrier integrity within the central nervous system. Cerebral malaria is a devastating disease. Patients have high levels of the protein HRPII in their blood. We have found that endothelial cell barriers become leaky when treated with concentrations of HRPII similar to those found in patients. This result suggests that HRPII may be important in cerebral malaria. Our finding that HRPII functions by causing inflammation suggests points of intervention for therapy or vaccination against this disease.
Collapse
|
183
|
Gasque P, Couderc T, Lecuit M, Roques P, Ng LFP. Chikungunya virus pathogenesis and immunity. Vector Borne Zoonotic Dis 2016; 15:241-9. [PMID: 25897810 DOI: 10.1089/vbz.2014.1710] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chikungunya virus (CHIKV) is an arbovirus associated with acute and chronic arthralgia that re-emerged in the Indian Ocean islands in 2005-2006 and is currently responsible for the ongoing outbreaks in the Caribbean islands and the Americas. We describe here the acute and chronic clinical manifestations of CHIKV in patients that define the disease. We also review the various animal models that have been developed to study CHIKV infection and pathology and further strengthened the understanding of the cellular and molecular mechanisms of CHIKV infection and immunity. A complete understanding of the immunopathogenesis of CHIKV infection will help develop the needed preventive and therapeutic approaches to combat this arbovirosis.
Collapse
Affiliation(s)
- Philippe Gasque
- 1 University of La Reunion , GRI/IRG EA4517, and Centre Hospitalier Universitaire (CHU North Felix-Guyon), Saint-Denis, La Reunion, France
| | | | | | | | | |
Collapse
|
184
|
Ahola T, Couderc T, Courderc T, Ng LFP, Hallengärd D, Powers A, Lecuit M, Esteban M, Merits A, Roques P, Liljeström P. Therapeutics and vaccines against chikungunya virus. Vector Borne Zoonotic Dis 2016; 15:250-7. [PMID: 25897811 DOI: 10.1089/vbz.2014.1681] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Currently, there are no licensed vaccines or therapies available against chikungunya virus (CHIKV), and these were subjects discussed during a CHIKV meeting recently organized in Langkawi, Malaysia. In this review, we chart the approaches taken in both areas. Because of a sharp increase in new data in these fields, the present paper is complementary to previous reviews by Weaver et al. in 2012 and Kaur and Chu in 2013 . The most promising antivirals so far discovered are reviewed, with a special focus on the virus-encoded replication proteins as potential targets. Within the vaccines in development, our review emphasizes the various strategies in parallel development that are unique in the vaccine field against a single disease.
Collapse
Affiliation(s)
- Tero Ahola
- 1 Department of Food and Environmental Sciences, University of Helsinki , Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Muthumani K, Block P, Flingai S, Muruganantham N, Chaaithanya IK, Tingey C, Wise M, Reuschel EL, Chung C, Muthumani A, Sarangan G, Srikanth P, Khan AS, Vijayachari P, Sardesai NY, Kim JJ, Ugen KE, Weiner DB. Rapid and Long-Term Immunity Elicited by DNA-Encoded Antibody Prophylaxis and DNA Vaccination Against Chikungunya Virus. J Infect Dis 2016; 214:369-78. [PMID: 27001960 PMCID: PMC4936642 DOI: 10.1093/infdis/jiw111] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 03/11/2016] [Indexed: 12/14/2022] Open
Abstract
Background. Vaccination and passive antibody therapies are critical for controlling infectious diseases. Passive antibody administration has limitations, including the necessity for purification and multiple injections for efficacy. Vaccination is associated with a lag phase before generation of immunity. Novel approaches reported here utilize the benefits of both methods for the rapid generation of effective immunity. Methods. A novel antibody-based prophylaxis/therapy entailing the electroporation-mediated delivery of synthetic DNA plasmids encoding biologically active anti–chikungunya virus (CHIKV) envelope monoclonal antibody (dMAb) was designed and evaluated for antiviral efficacy, as well as for the ability to overcome shortcomings inherent with conventional active vaccination and passive immunotherapy. Results. One intramuscular injection of dMAb produced antibodies in vivo more rapidly than active vaccination with an anti-CHIKV DNA vaccine. This dMAb neutralized diverse CHIKV clinical isolates and protected mice from viral challenge. Combination of dMAb and the CHIKV DNA vaccine afforded rapid and long-lived protection. Conclusions. A DNA-based dMAb strategy induced rapid protection against an emerging viral infection. This method can be combined with DNA vaccination as a novel strategy to provide both short- and long-term protection against this emerging infectious disease. These studies have implications for pathogen treatment and control strategies.
Collapse
Affiliation(s)
- Karuppiah Muthumani
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania The Wistar Institute, Philadelphia, Pennsylvania
| | - Peter Block
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania
| | - Seleeke Flingai
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania The Wistar Institute, Philadelphia, Pennsylvania
| | - Nagarajan Muruganantham
- Regional Medical Research Centers, Indian Council of Medical Research, Port Blair, Andaman & Nicobar Islands
| | - Itta Krishna Chaaithanya
- Regional Medical Research Centers, Indian Council of Medical Research, Port Blair, Andaman & Nicobar Islands
| | - Colleen Tingey
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania
| | - Megan Wise
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania The Wistar Institute, Philadelphia, Pennsylvania
| | - Emma L Reuschel
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania The Wistar Institute, Philadelphia, Pennsylvania
| | - Christopher Chung
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania The Wistar Institute, Philadelphia, Pennsylvania
| | - Abirami Muthumani
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania
| | - Gopalsamy Sarangan
- Department of Microbiology, Sri Ramachandra Medical College & Research Institute, Chennai, India
| | - Padma Srikanth
- Department of Microbiology, Sri Ramachandra Medical College & Research Institute, Chennai, India
| | - Amir S Khan
- Inovio Pharmaceutics Inc., Plymouth Meeting, Pennsylvania
| | - Paluru Vijayachari
- Regional Medical Research Centers, Indian Council of Medical Research, Port Blair, Andaman & Nicobar Islands
| | | | - J Joseph Kim
- Inovio Pharmaceutics Inc., Plymouth Meeting, Pennsylvania
| | - Kenneth E Ugen
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa
| | - David B Weiner
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania The Wistar Institute, Philadelphia, Pennsylvania
| |
Collapse
|
186
|
Adouchief S, Smura T, Sane J, Vapalahti O, Kurkela S. Sindbis virus as a human pathogen-epidemiology, clinical picture and pathogenesis. Rev Med Virol 2016; 26:221-41. [DOI: 10.1002/rmv.1876] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 02/02/2016] [Accepted: 02/04/2016] [Indexed: 01/24/2023]
Affiliation(s)
- Samuel Adouchief
- Department of Virology, Faculty of Medicine; University of Helsinki; Helsinki Finland
| | - Teemu Smura
- Department of Virology, Faculty of Medicine; University of Helsinki; Helsinki Finland
| | - Jussi Sane
- National institute for Health and Welfare (THL); Helsinki Finland
| | - Olli Vapalahti
- Department of Virology, Faculty of Medicine; University of Helsinki; Helsinki Finland
- Department of Virology and Immunology, HUSLAB; Helsinki University Central Hospital; Helsinki Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine; University of Helsinki; Helsinki Finland
| | - Satu Kurkela
- Department of Virology, Faculty of Medicine; University of Helsinki; Helsinki Finland
- Department of Virology and Immunology, HUSLAB; Helsinki University Central Hospital; Helsinki Finland
| |
Collapse
|
187
|
Furuyama W, Marzi A, Nanbo A, Haddock E, Maruyama J, Miyamoto H, Igarashi M, Yoshida R, Noyori O, Feldmann H, Takada A. Discovery of an antibody for pan-ebolavirus therapy. Sci Rep 2016; 6:20514. [PMID: 26861827 PMCID: PMC4748290 DOI: 10.1038/srep20514] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/07/2016] [Indexed: 01/19/2023] Open
Abstract
During the latest outbreak of Ebola virus disease in West Africa, monoclonal antibody therapy (e.g., ZMapp) was utilized to treat patients. However, due to the antigenic differences among the five ebolavirus species, the current therapeutic monoclonal antibodies are only effective against viruses of the species Zaire ebolavirus. Although this particular species has indeed caused the majority of human infections in Central and, recently, West Africa, other ebolavirus species (e.g., Sudan ebolavirus and Bundibugyo ebolavirus) have also repeatedly caused outbreaks in Central Africa and thus should not be neglected in the development of countermeasures against ebolaviruses. Here we report the generation of an ebolavirus glycoprotein-specific monoclonal antibody that effectively inhibits cellular entry of representative isolates of all known ebolavirus species in vitro and show its protective efficacy in mouse models of ebolavirus infections. This novel neutralizing monoclonal antibody targets a highly conserved internal fusion loop in the glycoprotein molecule and prevents membrane fusion of the viral envelope with cellular membranes. The discovery of this highly cross-neutralizing antibody provides a promising option for broad-acting ebolavirus antibody therapy and will accelerate the design of improved vaccines that can selectively elicit cross-neutralizing antibodies against multiple species of ebolaviruses.
Collapse
Affiliation(s)
- Wakako Furuyama
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana, USA
| | - Asuka Nanbo
- Department of Cell Physiology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Elaine Haddock
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana, USA
| | - Junki Maruyama
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Hiroko Miyamoto
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Manabu Igarashi
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Reiko Yoshida
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Osamu Noyori
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana, USA
| | - Ayato Takada
- Division of Global Epidemiology, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| |
Collapse
|
188
|
Jin J, Liss NM, Chen DH, Liao M, Fox JM, Shimak RM, Fong RH, Chafets D, Bakkour S, Keating S, Fomin ME, Muench MO, Sherman MB, Doranz BJ, Diamond MS, Simmons G. Neutralizing Monoclonal Antibodies Block Chikungunya Virus Entry and Release by Targeting an Epitope Critical to Viral Pathogenesis. Cell Rep 2015; 13:2553-2564. [PMID: 26686638 PMCID: PMC4720387 DOI: 10.1016/j.celrep.2015.11.043] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 10/15/2015] [Accepted: 11/11/2015] [Indexed: 11/26/2022] Open
Abstract
We evaluated the mechanism by which neutralizing human monoclonal antibodies inhibit chikungunya virus (CHIKV) infection. Potently neutralizing antibodies (NAbs) blocked infection at multiple steps of the virus life cycle, including entry and release. Cryo-electron microscopy structures of Fab fragments of two human NAbs and chikungunya virus-like particles showed a binding footprint that spanned independent domains on neighboring E2 subunits within one viral spike, suggesting a mechanism for inhibiting low-pH-dependent membrane fusion. Detailed epitope mapping identified amino acid E2-W64 as a critical interaction residue. An escape mutation (E2-W64G) at this residue rendered CHIKV attenuated in mice. Consistent with these data, CHIKV-E2-W64G failed to emerge in vivo under the selection pressure of one of the NAbs, IM-CKV063. As our study suggests that antibodies engaging the residue E2-W64 can potently inhibit CHIKV at multiple stages of infection, antibody-based therapies or immunogens that target this region might have protective value.
Collapse
Affiliation(s)
- Jing Jin
- Blood Systems Research Institute, San Francisco, CA 94118, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nathan M Liss
- Blood Systems Research Institute, San Francisco, CA 94118, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dong-Hua Chen
- Department of Structural Biology, Stanford University, Stanford, CA 94305, USA
| | - Maofu Liao
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Julie M Fox
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Raeann M Shimak
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rachel H Fong
- Integral Molecular Inc., Philadelphia, PA 19104, USA
| | - Daniel Chafets
- Blood Systems Research Institute, San Francisco, CA 94118, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sonia Bakkour
- Blood Systems Research Institute, San Francisco, CA 94118, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sheila Keating
- Blood Systems Research Institute, San Francisco, CA 94118, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marina E Fomin
- Blood Systems Research Institute, San Francisco, CA 94118, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marcus O Muench
- Blood Systems Research Institute, San Francisco, CA 94118, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael B Sherman
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Graham Simmons
- Blood Systems Research Institute, San Francisco, CA 94118, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
189
|
Madariaga M, Ticona E, Resurrecion C. Chikungunya: bending over the Americas and the rest of the world. Braz J Infect Dis 2015; 20:91-8. [PMID: 26707971 PMCID: PMC9425360 DOI: 10.1016/j.bjid.2015.10.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 10/13/2015] [Accepted: 10/16/2015] [Indexed: 11/25/2022] Open
Abstract
Chikungunya is an arthropod-borne virus transmitted by Aedes mosquito bites. A viral mutation has allowed Aedes albopictus to become the preferred vector extending the geographic spread of the condition. The virus causes an acute febrile illness occasionally followed by a chronic rheumatic condition causing severe impairment. The diagnosis is usually confirmed with serology. No specific treatment is currently available. This article reviews the condition with emphasis on his dissemination in the Americas.
Collapse
Affiliation(s)
- Miguel Madariaga
- Section of Infectious Diseases, Naples Community Hospital, Naples, United States.
| | - Eduardo Ticona
- Hospital Nacional Dos de Mayo, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | | |
Collapse
|
190
|
Long F, Fong RH, Austin SK, Chen Z, Klose T, Fokine A, Liu Y, Porta J, Sapparapu G, Akahata W, Doranz BJ, Crowe JE, Diamond MS, Rossmann MG. Cryo-EM structures elucidate neutralizing mechanisms of anti-chikungunya human monoclonal antibodies with therapeutic activity. Proc Natl Acad Sci U S A 2015; 112:13898-903. [PMID: 26504196 PMCID: PMC4653152 DOI: 10.1073/pnas.1515558112] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that causes severe acute and chronic disease in humans. Although highly inhibitory murine and human monoclonal antibodies (mAbs) have been generated, the structural basis of their neutralizing activity remains poorly characterized. Here, we determined the cryo-EM structures of chikungunya virus-like particles complexed with antibody fragments (Fab) of two highly protective human mAbs, 4J21 and 5M16, that block virus fusion with host membranes. Both mAbs bind primarily to sites within the A and B domains, as well as to the B domain's β-ribbon connector of the viral glycoprotein E2. The footprints of these antibodies on the viral surface were consistent with results from loss-of-binding studies using an alanine scanning mutagenesis-based epitope mapping approach. The Fab fragments stabilized the position of the B domain relative to the virus, particularly for the complex with 5M16. This finding is consistent with a mechanism of neutralization in which anti-CHIKV mAbs that bridge the A and B domains impede movement of the B domain away from the underlying fusion loop on the E1 glycoprotein and therefore block the requisite pH-dependent fusion of viral and host membranes.
Collapse
Affiliation(s)
- Feng Long
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | | | - Stephen K Austin
- Departments of Medicine, Molecular Microbiology, Pathology, and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Zhenguo Chen
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Thomas Klose
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Andrei Fokine
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Yue Liu
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Jason Porta
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907
| | - Gopal Sapparapu
- Department of Pediatrics, Vanderbilt University, Nashville, TN 37232
| | | | | | - James E Crowe
- Department of Pediatrics, Vanderbilt University, Nashville, TN 37232; Departments of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232; Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology, and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael G Rossmann
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907;
| |
Collapse
|
191
|
Fox JM, Long F, Edeling MA, Lin H, van Duijl-Richter MKS, Fong RH, Kahle KM, Smit JM, Jin J, Simmons G, Doranz BJ, Crowe JE, Fremont DH, Rossmann MG, Diamond MS. Broadly Neutralizing Alphavirus Antibodies Bind an Epitope on E2 and Inhibit Entry and Egress. Cell 2015; 163:1095-1107. [PMID: 26553503 DOI: 10.1016/j.cell.2015.10.050] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/18/2015] [Accepted: 10/19/2015] [Indexed: 01/12/2023]
Abstract
We screened a panel of mouse and human monoclonal antibodies (MAbs) against chikungunya virus and identified several with inhibitory activity against multiple alphaviruses. Passive transfer of broadly neutralizing MAbs protected mice against infection by chikungunya, Mayaro, and O'nyong'nyong alphaviruses. Using alanine-scanning mutagenesis, loss-of-function recombinant proteins and viruses, and multiple functional assays, we determined that broadly neutralizing MAbs block multiple steps in the viral lifecycle, including entry and egress, and bind to a conserved epitope on the B domain of the E2 glycoprotein. A 16 Å resolution cryo-electron microscopy structure of a Fab fragment bound to CHIKV E2 B domain provided an explanation for its neutralizing activity. Binding to the B domain was associated with repositioning of the A domain of E2 that enabled cross-linking of neighboring spikes. Our results suggest that B domain antigenic determinants could be targeted for vaccine or antibody therapeutic development against multiple alphaviruses of global concern.
Collapse
Affiliation(s)
- Julie M Fox
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Feng Long
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Melissa A Edeling
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hueylie Lin
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Rachel H Fong
- Integral Molecular, Inc., Philadelphia, PA 19104, USA
| | | | - Jolanda M Smit
- University of Groningen and University Medical Center Groningen, 9713 GZ Groningen, the Netherlands
| | - Jing Jin
- Blood Systems Research Institute, San Francisco, CA 94118, USA
| | - Graham Simmons
- Blood Systems Research Institute, San Francisco, CA 94118, USA
| | | | - James E Crowe
- Departments of Pediatrics, Pathology, Microbiology, and Immunology and the Vanderbilt Vaccine Center, Vanderbilt University, Nashville, TN 37235, USA
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael G Rossmann
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
192
|
Erasmus JH, Needham J, Raychaudhuri S, Diamond MS, Beasley DWC, Morkowski S, Salje H, Fernandez Salas I, Kim DY, Frolov I, Nasar F, Weaver SC. Utilization of an Eilat Virus-Based Chimera for Serological Detection of Chikungunya Infection. PLoS Negl Trop Dis 2015; 9:e0004119. [PMID: 26492074 PMCID: PMC4619601 DOI: 10.1371/journal.pntd.0004119] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/04/2015] [Indexed: 12/16/2022] Open
Abstract
In December of 2013, chikungunya virus (CHIKV), an alphavirus in the family Togaviridae, was introduced to the island of Saint Martin in the Caribbean, resulting in the first autochthonous cases reported in the Americas. As of January 2015, local and imported CHIKV has been reported in 50 American countries with over 1.1 million suspected cases. CHIKV causes a severe arthralgic disease for which there are no approved vaccines or therapeutics. Furthermore, the lack of a commercially available, sensitive, and affordable diagnostic assay limits surveillance and control efforts. To address this issue, we utilized an insect-specific alphavirus, Eilat virus (EILV), to develop a diagnostic antigen that does not require biosafety containment facilities to produce. We demonstrated that EILV/CHIKV replicates to high titers in insect cells and can be applied directly in enzyme-linked immunosorbent assays without inactivation, resulting in highly sensitive detection of recent and past CHIKV infection, and outperforming traditional antigen preparations. We have developed an innovative approach to production of alphavirus antigens for use in diagnostic assays that results in reduced production complexity as well as improved sensitivity in application. By generating recombinant viruses that contain the structural protein genes of pathogenic alphaviruses and the nonstructural protein genes of an insect-specific alphavirus, Eilat virus, we have been able to produce insect-restricted viruses that are antigenically identical to their pathogenic counterparts. The insect-specific nature of these chimeric viruses yields an advantageous safety profile and allows for safe handling of the antigen at the bench top. Traditional antigens, produced from wild-type virus, require extensive processing, from growth at biosafety level 3 to concentration and inactivation, followed by lyophilization, which often compromises antigen reactivity and is financially costly. Furthermore, current inactivation methods are imperfect processes that have historically resulted in residual live virus and subsequent breach of containment when used in a diagnostic setting. Other approaches, such as recombinant antigens generated from viral particle subunits, are missing conformational epitopes and their application results in reduced sensitivity. Here we describe the development of a diagnostic assay using this technology for the detection of chikungunya infection in humans.
Collapse
Affiliation(s)
- Jesse H. Erasmus
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Tropical Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - James Needham
- InBios International, Inc., Seattle, Washington, United States of America
| | | | - Michael S. Diamond
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - David W. C. Beasley
- Institute for Human Infections and Immunity, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Biodefense and Emerging Infectious Diseases, and Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Stan Morkowski
- InBios International, Inc., Seattle, Washington, United States of America
| | - Henrik Salje
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Pasteur Institute, Paris, France
| | | | - Dal Young Kim
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Ilya Frolov
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Farooq Nasar
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Tropical Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Scott C. Weaver
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Tropical Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute for Human Infections and Immunity, Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
193
|
Nonhuman Primate Models of Chikungunya Virus Infection and Disease (CHIKV NHP Model). Pathogens 2015; 4:662-81. [PMID: 26389957 PMCID: PMC4584280 DOI: 10.3390/pathogens4030662] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/04/2015] [Accepted: 09/09/2015] [Indexed: 01/18/2023] Open
Abstract
Chikungunya virus (CHIKV) is a positive-sense RNA virus transmitted by Aedes mosquitoes. CHIKV is a reemerging Alphavirus that causes acute febrile illness and severe and debilitating polyarthralgia of the peripheral joints. Huge epidemics and the rapid spread of CHIKV seen in India and the Indian Ocean region established CHIKV as a global health concern. This concern was further solidified by the recent incursion of the virus into the Western hemisphere, a region without pre-existing immunity. Nonhuman primates (NHPs) serve as excellent animal models for understanding CHIKV pathogenesis and pre-clinical assessment of vaccines and therapeutics. NHPs present advantages over rodent models because they are a natural amplification host for CHIKV and they share significant genetic and physiological homology with humans. CHIKV infection in NHPs results in acute fever, rash, viremia and production of type I interferon. NHPs develop CHIKV-specific B and T-cells, generating neutralizing antibodies and CHIKV-specific CD4⁺ and CD8⁺ T-cells. CHIKV establishes a persistent infection in NHPs, particularly in cynomolgus macaques, because infectious virus could be recovered from spleen, liver, and muscle as late as 44 days post infection. NHPs are valuable models that are useful in preclinical testing of vaccines and therapeutics and uncovering the details of CHIKV pathogenesis.
Collapse
|
194
|
Defining New Therapeutics Using a More Immunocompetent Mouse Model of Antibody-Enhanced Dengue Virus Infection. mBio 2015; 6:e01316-15. [PMID: 26374123 PMCID: PMC4600115 DOI: 10.1128/mbio.01316-15] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
With over 3.5 billion people at risk and approximately 390 million human infections per year, dengue virus (DENV) disease strains health care resources worldwide. Previously, we and others established models for DENV pathogenesis in mice that completely lack subunits of the receptors (Ifnar and Ifngr) for type I and type II interferon (IFN) signaling; however, the utility of these models is limited by the pleotropic effect of these cytokines on innate and adaptive immune system development and function. Here, we demonstrate that the specific deletion of Ifnar expression on subsets of murine myeloid cells (LysM Cre+Ifnarflox/flox [denoted as Ifnarf/f herein]) resulted in enhanced DENV replication in vivo. The administration of subneutralizing amounts of cross-reactive anti-DENV monoclonal antibodies to LysM Cre+Ifnarf/f mice prior to infection with DENV serotype 2 or 3 resulted in antibody-dependent enhancement (ADE) of infection with many of the characteristics associated with severe DENV disease in humans, including plasma leakage, hypercytokinemia, liver injury, hemoconcentration, and thrombocytopenia. Notably, the pathogenesis of severe DENV-2 or DENV-3 infection in LysM Cre+Ifnarf/f mice was blocked by pre- or postexposure administration of a bispecific dual-affinity retargeting molecule (DART) or an optimized RIG-I receptor agonist that stimulates innate immune responses. Our findings establish a more immunocompetent animal model of ADE of infection with multiple DENV serotypes in which disease is inhibited by treatment with broad-spectrum antibody derivatives or innate immune stimulatory agents. Although dengue virus (DENV) infects hundreds of millions of people annually and results in morbidity and mortality on a global scale, there are no approved antiviral treatments or vaccines. Part of the difficulty in evaluating therapeutic candidates is the lack of small animal models that are permissive to DENV and recapitulate the clinical features of severe human disease. Using animals lacking the type I interferon receptor only on myeloid cell subsets, we developed a more immunocompetent mouse model of severe DENV infection with characteristics of the human disease, including vascular leakage, hemoconcentration, thrombocytopenia, and liver injury. Using this model, we demonstrate that pathogenesis by two different DENV serotypes is inhibited by therapeutic administration of a genetically modified antibody or a RIG-I receptor agonist that stimulates innate immunity.
Collapse
|
195
|
Mota MTDO, Ribeiro MR, Vedovello D, Nogueira ML. Mayaro virus: a neglected arbovirus of the Americas. Future Virol 2015. [DOI: 10.2217/fvl.15.76] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Mayaro virus is a neglected tropical arbovirus that causes a mild, self-limited febrile syndrome, sometimes accompanied by a highly incapacitating arthralgia. First isolated in Trinidad and Tobago in 1954, it was reported in several countries within the tropical regions of South and Central America. Human infections are accidental spillover of the enzootic cycle. Little epidemiological data are available due to inadequate surveillance and the generic nature of clinical manifestations resulting in the misdiagnosis with other viral fevers. Despite its restricted distribution, Mayaro fever may become a public health issue due to their urbanization potential. Accurate epidemiological data are urgently needed to access the real distribution of this virus guiding public health policies better.
Collapse
Affiliation(s)
- Mânlio Tasso de Oliveira Mota
- Faculty of Medicine of São José do Rio Preto (FAMERP),5416 Brigadeiro Faria Lima Avenue, São José do Rio Preto, SP, Brazil, 15090-000
| | - Milene Rocha Ribeiro
- Faculty of Medicine of São José do Rio Preto (FAMERP),5416 Brigadeiro Faria Lima Avenue, São José do Rio Preto, SP, Brazil, 15090-000
| | - Danila Vedovello
- Faculty of Medicine of São José do Rio Preto (FAMERP),5416 Brigadeiro Faria Lima Avenue, São José do Rio Preto, SP, Brazil, 15090-000
| | - Maurício Lacerda Nogueira
- Faculty of Medicine of São José do Rio Preto (FAMERP),5416 Brigadeiro Faria Lima Avenue, São José do Rio Preto, SP, Brazil, 15090-000
| |
Collapse
|
196
|
Abdelnabi R, Neyts J, Delang L. Towards antivirals against chikungunya virus. Antiviral Res 2015; 121:59-68. [PMID: 26119058 PMCID: PMC7113767 DOI: 10.1016/j.antiviral.2015.06.017] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/18/2015] [Accepted: 06/24/2015] [Indexed: 12/25/2022]
Abstract
Chikungunya virus (CHIKV) has re-emerged in recent decades, causing major outbreaks of chikungunya fever in many parts of Africa and Asia, and since the end of 2013 also in Central and South America. Infections are usually associated with a low mortality rate, but can proceed into a painful chronic stage, during which patients may suffer from polyarthralgia and joint stiffness for weeks and even several years. There are no vaccines or antiviral drugs available for the prevention or treatment of CHIKV infections. Current therapy therefore consists solely of the administration of analgesics, antipyretics and anti-inflammatory agents to relieve symptoms. We here review molecules that have been reported to inhibit CHIKV replication, either as direct-acting antivirals, host-targeting drugs or those that act via a yet unknown mechanism. This article forms part of a symposium in Antiviral Research on "Chikungunya discovers the New World."
Collapse
Affiliation(s)
- Rana Abdelnabi
- Rega Institute for Medical Research, University of Leuven, Belgium
| | - Johan Neyts
- Rega Institute for Medical Research, University of Leuven, Belgium.
| | - Leen Delang
- Rega Institute for Medical Research, University of Leuven, Belgium
| |
Collapse
|
197
|
Lam S, Nyo M, Phuektes P, Yew CW, Tan YJ, Chu JJH. A potent neutralizing IgM mAb targeting the N218 epitope on E2 protein protects against Chikungunya virus pathogenesis. MAbs 2015; 7:1178-94. [PMID: 26305993 DOI: 10.1080/19420862.2015.1083664] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Chikungunya virus (CHIKV) is a medically important human viral pathogen that causes Chikungunya fever accompanied with debilitating and persistent joint pain. Host-elicited or passively-transferred monoclonal antibodies (mAb) are essential mediators of CHIKV clearance. Therefore, this study aimed to generate and characterize a panel of mAbs for their neutralization efficacy against CHIKV infection in a cell-based and murine model. To evaluate their antigenicity and neutralization profile, indirect enzyme-linked immunosorbent assay (ELISA), an immunofluorescence assay (IFA) and a plaque reduction neutralization test were performed on mAbs of IgM isotype. CHIKV escape mutants against mAb 3E7b neutralization were generated, and reverse genetics techniques were then used to create an infectious CHIKV clone with a single mutation. 3E7b was also administered to neonate mice prior or after CHIKV infection. The survival rate, CHIKV burden in tissues and histopathology of the limb muscles were evaluated. Both IgM 3E7b and 8A2c bind strongly to native CHIKV surface and potently neutralize CHIKV replication. Further analyses of 3E7b binding and neutralization of CHIKV single-mutant clones revealed that N218 of CHIKV E2 protein is a potent neutralizing epitope. In a pre-binding neutralization assay, 3E7b blocks CHIKV attachment to permissive cells, possibly by binding to the surface-accessible E2-N218 residue. Prophylactic administration of 3E7b to neonate mice markedly reduced viremia and protected against CHIKV pathogenesis in various mice tissues. Given therapeutically at 4 h post-infection, 3E7b conferred 100% survival rate and similarly reduced CHIKV load in most mice tissues except the limb muscles. Collectively, these findings highlight the usefulness of 3E7b for future prophylactic or epitope-based vaccine design.
Collapse
Affiliation(s)
- Shirley Lam
- a Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology; Yong Loo Lin School of Medicine, National University Health System, National University of Singapore ; Singapore
| | - Min Nyo
- a Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology; Yong Loo Lin School of Medicine, National University Health System, National University of Singapore ; Singapore
| | - Patchara Phuektes
- a Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology; Yong Loo Lin School of Medicine, National University Health System, National University of Singapore ; Singapore
| | - Chow Wenn Yew
- b Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research) ; Singapore
| | - Yee Joo Tan
- b Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research) ; Singapore.,c Hepatitis Viruses and Newly Emerging Viruses; Department of Microbiology; Yong Loo Lin School of Medicine, National University Health System, National University of Singapore ; Singapore
| | - Justin Jang Hann Chu
- a Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology; Yong Loo Lin School of Medicine, National University Health System, National University of Singapore ; Singapore.,b Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research) ; Singapore
| |
Collapse
|
198
|
Lum FM, Ng LF. Cellular and molecular mechanisms of chikungunya pathogenesis. Antiviral Res 2015; 120:165-74. [DOI: 10.1016/j.antiviral.2015.06.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 05/27/2015] [Accepted: 06/16/2015] [Indexed: 12/15/2022]
|
199
|
Stapleford KA, Coffey LL, Lay S, Bordería AV, Duong V, Isakov O, Rozen-Gagnon K, Arias-Goeta C, Blanc H, Beaucourt S, Haliloğlu T, Schmitt C, Bonne I, Ben-Tal N, Shomron N, Failloux AB, Buchy P, Vignuzzi M. Emergence and transmission of arbovirus evolutionary intermediates with epidemic potential. Cell Host Microbe 2015; 15:706-16. [PMID: 24922573 DOI: 10.1016/j.chom.2014.05.008] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 03/19/2014] [Accepted: 04/22/2014] [Indexed: 01/14/2023]
Abstract
The high replication and mutation rates of RNA viruses can result in the emergence of new epidemic variants. Thus, the ability to follow host-specific evolutionary trajectories of viruses is essential to predict and prevent epidemics. By studying the spatial and temporal evolution of chikungunya virus during natural transmission between mosquitoes and mammals, we have identified viral evolutionary intermediates prior to emergence. Analysis of virus populations at anatomical barriers revealed that the mosquito midgut and salivary gland pose population bottlenecks. By focusing on virus subpopulations in the saliva of multiple mosquito strains, we recapitulated the emergence of a recent epidemic strain of chikungunya and identified E1 glycoprotein mutations with potential to emerge in the future. These mutations confer fitness advantages in mosquito and mammalian hosts by altering virion stability and fusogenic activity. Thus, virus evolutionary trajectories can be predicted and studied in the short term before new variants displace currently circulating strains.
Collapse
Affiliation(s)
- Kenneth A Stapleford
- Viral Populations and Pathogenesis Unit, Institut Pasteur, Centre National de la Recherche Scientifique UMR 3569, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Lark L Coffey
- Center for Vectorborne Diseases, Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, One Shields Avenue, 5327 VM3A, Davis, CA 95616, USA
| | - Sreyrath Lay
- Virology Unit, Institut Pasteur in Cambodia 5, Monivong Boulevard, PO Box 983, Phnom Penh, Cambodia
| | - Antonio V Bordería
- Viral Populations and Pathogenesis Unit, Institut Pasteur, Centre National de la Recherche Scientifique UMR 3569, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Veasna Duong
- Virology Unit, Institut Pasteur in Cambodia 5, Monivong Boulevard, PO Box 983, Phnom Penh, Cambodia
| | - Ofer Isakov
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Kathryn Rozen-Gagnon
- Viral Populations and Pathogenesis Unit, Institut Pasteur, Centre National de la Recherche Scientifique UMR 3569, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Camilo Arias-Goeta
- Arboviruses and Insect Vectors Lab, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Hervé Blanc
- Viral Populations and Pathogenesis Unit, Institut Pasteur, Centre National de la Recherche Scientifique UMR 3569, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Stéphanie Beaucourt
- Viral Populations and Pathogenesis Unit, Institut Pasteur, Centre National de la Recherche Scientifique UMR 3569, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Türkan Haliloğlu
- Department of Chemical Engineering and Polymer Research Center, Boğaziçi University, Bebek 34342, Istanbul, Turkey
| | - Christine Schmitt
- Ultrastructural Microscopy Platform, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Isabelle Bonne
- Ultrastructural Microscopy Platform, Institut Pasteur, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Nir Ben-Tal
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Noam Shomron
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anna-Bella Failloux
- Arboviruses and Insect Vectors Lab, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Philippe Buchy
- Virology Unit, Institut Pasteur in Cambodia 5, Monivong Boulevard, PO Box 983, Phnom Penh, Cambodia
| | - Marco Vignuzzi
- Viral Populations and Pathogenesis Unit, Institut Pasteur, Centre National de la Recherche Scientifique UMR 3569, 28 rue du Dr Roux, 75724 Paris Cedex 15, France.
| |
Collapse
|
200
|
Smith SA, Silva LA, Fox JM, Flyak AI, Kose N, Sapparapu G, Khomandiak S, Khomadiak S, Ashbrook AW, Kahle KM, Fong RH, Swayne S, Doranz BJ, McGee CE, Heise MT, Pal P, Brien JD, Austin SK, Diamond MS, Dermody TS, Crowe JE. Isolation and Characterization of Broad and Ultrapotent Human Monoclonal Antibodies with Therapeutic Activity against Chikungunya Virus. Cell Host Microbe 2015; 18:86-95. [PMID: 26159721 PMCID: PMC4501771 DOI: 10.1016/j.chom.2015.06.009] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/27/2015] [Accepted: 06/22/2015] [Indexed: 11/25/2022]
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted RNA virus that causes acute febrile infection associated with polyarthralgia in humans. Mechanisms of protective immunity against CHIKV are poorly understood, and no effective therapeutics or vaccines are available. We isolated and characterized human monoclonal antibodies (mAbs) that neutralize CHIKV infectivity. Among the 30 mAbs isolated, 13 had broad and ultrapotent neutralizing activity (IC50 < 10 ng/ml), and all of these mapped to domain A of the E2 envelope protein. Potent inhibitory mAbs blocked post-attachment steps required for CHIKV membrane fusion, and several were protective in a lethal challenge model in immunocompromised mice, even when administered at late time points after infection. These highly protective mAbs could be considered for prevention or treatment of CHIKV infection, and their epitope location in domain A of E2 could be targeted for rational structure-based vaccine development.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/isolation & purification
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/isolation & purification
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Viral/immunology
- Antibodies, Viral/isolation & purification
- Antibodies, Viral/therapeutic use
- Chemoprevention/methods
- Chikungunya Fever/therapy
- Chikungunya virus/immunology
- Chikungunya virus/physiology
- Disease Models, Animal
- Humans
- Immunization, Passive/methods
- Inhibitory Concentration 50
- Mice
- Protein Binding
- Survival Analysis
- Treatment Outcome
- Viral Envelope Proteins/immunology
- Virus Internalization/drug effects
Collapse
Affiliation(s)
- Scott A Smith
- Department of Medicine, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Laurie A Silva
- Department of Pediatrics, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Julie M Fox
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew I Flyak
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Nurgun Kose
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Gopal Sapparapu
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | | | - Solomiia Khomadiak
- Department of Pediatrics, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Alison W Ashbrook
- Department of Pediatrics, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | | | | | | | | | - Charles E McGee
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Mark T Heise
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Pankaj Pal
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James D Brien
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - S Kyle Austin
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Terence S Dermody
- Department of Pediatrics, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA
| | - James E Crowe
- Department of Pediatrics, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|