201
|
HUANG Z, DENG N, YAN GQ, GAO MX, LIANG Z, ZHANG LH, ZHANG XM, ZHANG YK. Array-Based Two Dimensional Liquid Chromatography System for Proteomic Analysis of Human Plasma. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2015. [DOI: 10.1016/s1872-2040(15)60865-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
202
|
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
|
203
|
Wei Chiam C, Fun Chan Y, Chai Ong K, Thong Wong K, Sam IC. Neurovirulence comparison of chikungunya virus isolates of the Asian and East/Central/South African genotypes from Malaysia. J Gen Virol 2015; 96:3243-3254. [PMID: 26276497 DOI: 10.1099/jgv.0.000263] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Chikungunya virus (CHIKV), an alphavirus of the family Togaviridae, causes fever, polyarthritis and rash. There are three genotypes: West African, Asian and East/Central/South African (ECSA). The latter two genotypes have caused global outbreaks in recent years. Recent ECSA CHIKV outbreaks have been associated with severe neurological disease, but it is not known if different CHIKV genotypes are associated with different neurovirulence. In this study, the neurovirulence of Asian (MY/06/37348) and ECSA (MY/08/065) strains of CHIKV isolated in Malaysia were compared. Intracerebral inoculation of either virus into suckling mice was followed by virus titration, histopathology and gene expression analysis of the harvested brains. Both strains of CHIKV replicated similarly, yet mice infected with MY/06/37348 showed higher mortality. Histopathology findings showed that both CHIKV strains spread within the brain (where CHIKV antigen was localized to astrocytes and neurons) and beyond to skeletal muscle. In MY/06/37348-infected mice, apoptosis, which is associated with neurovirulence in alphaviruses, was observed earlier in brains. Comparison of gene expression showed that a pro-apoptotic gene (eIF2αK2) was upregulated at higher levels in MY/06/37348-infected mice, while genes involved in anti-apoptosis (BIRC3), antiviral responses and central nervous system protection (including CD40, IL-10RA, MyD88 and PYCARD) were upregulated more highly in MY/08/065-infected mice. In conclusion, the higher mortality observed following MY/06/37348 infection in mice is due not to higher viral replication in the brain, but to differentially expressed genes involved in host immune responses. These findings may help to identify therapeutic strategies and biomarkers for neurological CHIKV infections.
Collapse
Affiliation(s)
- Chun Wei Chiam
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Yoke Fun Chan
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Kien Chai Ong
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Kum Thong Wong
- Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - I-Ching Sam
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| |
Collapse
|
204
|
Limitations of Current in Vivo Mouse Models for the Study of Chikungunya Virus Pathogenesis. Med Sci (Basel) 2015; 3:64-77. [PMID: 29083392 PMCID: PMC5635755 DOI: 10.3390/medsci3030064] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/20/2015] [Accepted: 07/20/2015] [Indexed: 12/19/2022] Open
Abstract
Chikungunya virus (CHIKV) is an arthropod-borne alphavirus that causes febrile chikungunya fever (CHIKF) in humans. This disease is debilitating and characterized by acute fever onset and chronic incapacitating polyarthralgia. CHIKF pathogenesis remains poorly defined with no approved vaccines and therapies. Recent outbreaks in the Caribbean islands have elevated concerns over the possibility of a global pandemic. Tremendous efforts have been made to develop relevant mouse models to enable the study of infection and immunity against this viral disease. Among them, the more common C57BL/6 mouse model demonstrated the ability to recapitulate the symptoms shown in infected humans, including self-limiting arthritis, myositis, and tenosynovitis. This has facilitated the unraveling of some key factors involved in disease pathogenesis of CHIKF. However, the stark differences in immune response between humans and mouse models necessitate the development of an animal model with an immune system that is more genetically similar to the human system for a better representation. In this paper, we aim to uncover the limitations of the C57BL/6 model and discuss alternative mouse models for CHIKV research.
Collapse
|
205
|
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]
|
206
|
Her Z, Teng TS, Tan JJL, Teo TH, Kam YW, Lum FM, Lee WWL, Gabriel C, Melchiotti R, Andiappan AK, Lulla V, Lulla A, Win MK, Chow A, Biswas SK, Leo YS, Lecuit M, Merits A, Rénia L, Ng LFP. Loss of TLR3 aggravates CHIKV replication and pathology due to an altered virus-specific neutralizing antibody response. EMBO Mol Med 2015; 7:24-41. [PMID: 25452586 PMCID: PMC4309666 DOI: 10.15252/emmm.201404459] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
RNA-sensing toll-like receptors (TLRs) mediate innate immunity and regulate anti-viral response. We show here that TLR3 regulates host immunity and the loss of TLR3 aggravates pathology in Chikungunya virus (CHIKV) infection. Susceptibility to CHIKV infection is markedly increased in human and mouse fibroblasts with defective TLR3 signaling. Up to 100-fold increase in CHIKV load was observed in Tlr3−/− mice, alongside increased virus dissemination and pro-inflammatory myeloid cells infiltration. Infection in bone marrow chimeric mice showed that TLR3-expressing hematopoietic cells are required for effective CHIKV clearance. CHIKV-specific antibodies from Tlr3−/− mice exhibited significantly lower in vitro neutralization capacity, due to altered virus-neutralizing epitope specificity. Finally, SNP genotyping analysis of CHIKF patients on TLR3 identified SNP rs6552950 to be associated with disease severity and CHIKV-specific neutralizing antibody response. These results demonstrate a key role for TLR3-mediated antibody response to CHIKV infection, virus replication and pathology, providing a basis for future development of immunotherapeutics in vaccine development.
Collapse
Affiliation(s)
- Zhisheng Her
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Terk-Shin Teng
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Jeslin J L Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Teck-Hui Teo
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
| | - Yiu-Wing Kam
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Fok-Moon Lum
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wendy W L Lee
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
| | - Christelle Gabriel
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Rossella Melchiotti
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore Doctoral School in Translational and Molecular Medicine (DIMET), University of Milano-Bicocca, Milan, Italy
| | - Anand K Andiappan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Valeria Lulla
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Aleksei Lulla
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Mar K Win
- Institute of Infectious Disease and Epidemiology (IIDE), Tan Tock Seng Hospital, Singapore, Singapore
| | - Angela Chow
- Institute of Infectious Disease and Epidemiology (IIDE), Tan Tock Seng Hospital, Singapore, Singapore
| | - Subhra K Biswas
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Yee-Sin Leo
- Institute of Infectious Disease and Epidemiology (IIDE), Tan Tock Seng Hospital, Singapore, Singapore
| | - Marc Lecuit
- Institut Pasteur, Biology of Infection Unit, Paris, France Inserm U1117, Paris, France Paris Descartes University Sorbonne Paris Cité, Necker-Enfants Malades University Hospital, Institut Imagine, Paris, France
| | - Andres Merits
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Laurent Rénia
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Lisa F P Ng
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| |
Collapse
|
207
|
Abstract
Chikungunya virus (CHIKV) is a rapidly emerging mosquito-borne alphavirus causing millions of infections in the tropical and subtropical regions of the world. CHIKV infection often leads to an acute self-limited febrile illness with debilitating myalgia and arthralgia. A potential long-term complication of CHIKV infection is severe joint pain, which can last for months to years. There are no vaccines or specific therapeutics available to prevent or treat infection. This review describes the critical steps in CHIKV cell entry. We summarize the latest studies on the virus-cell tropism, virus-receptor binding, internalization, membrane fusion and review the molecules and compounds that have been described to interfere with virus cell entry. The aim of the review is to give the reader a state-of-the-art overview on CHIKV cell entry and to provide an outlook on potential new avenues in CHIKV research.
Collapse
|
208
|
Couderc T, Lecuit M. Chikungunya virus pathogenesis: From bedside to bench. Antiviral Res 2015; 121:120-31. [PMID: 26159730 DOI: 10.1016/j.antiviral.2015.07.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/04/2015] [Indexed: 11/28/2022]
Abstract
Chikungunya virus (CHIKV) is an arbovirus transmitted to humans by mosquito bite. A decade ago, the virus caused a major outbreak in the islands of the Indian Ocean, then reached India and Southeast Asia. More recently, CHIKV has emerged in the Americas, first reaching the Caribbean and now extending to Central, South and North America. It is therefore considered a major public health and economic threat. CHIKV causes febrile illness typically associated with debilitating joint pains. In rare cases, it may also cause central nervous system disease, notably in neonates. Joint symptoms may persist for months to years, and lead to arthritis. This review focuses on the spectrum of signs and symptoms associated with CHIKV infection in humans. It also illustrates how the analysis of clinical and biological data from human cohorts and the development of animal and cellular models of infection has helped to identify the tissue and cell tropisms of the virus and to decipher host responses in benign, severe or persistent disease. This article forms part of a symposium in Antiviral Research on "Chikungunya discovers the New World".
Collapse
Affiliation(s)
- Thérèse Couderc
- Institut Pasteur, Biology of Infection Unit, Paris, France; Inserm U1117, Paris, France.
| | - Marc Lecuit
- Institut Pasteur, Biology of Infection Unit, Paris, France; Inserm U1117, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Division of Infectious Diseases and Tropical Medicine, Necker-Enfants Malades University Hospital, Institut Imagine, Paris, France; Global Virus Network.
| |
Collapse
|
209
|
A Rodent Model of Chikungunya Virus Infection in RAG1 -/- Mice, with Features of Persistence, for Vaccine Safety Evaluation. PLoS Negl Trop Dis 2015; 9:e0003800. [PMID: 26115459 PMCID: PMC4482609 DOI: 10.1371/journal.pntd.0003800] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 04/28/2015] [Indexed: 12/23/2022] Open
Abstract
Chikungunya virus (CHIKV) is a positive sense, single stranded RNA virus in the genus Alphavirus, and the etiologic agent of epidemics of severe arthralgia in Africa, Asia, Europe and, most recently, the Americas. CHIKV causes chikungunya fever (CHIK), a syndrome characterized by rash, fever, and debilitating, often chronic arthritis. In recent outbreaks, CHIKV has been recognized to manifest more neurologic signs of illness in the elderly and those with co-morbidities. The syndrome caused by CHIKV is often self-limited; however, many patients develop persistent arthralgia that can last for months or years. These characteristics make CHIKV not only important from a human health standpoint, but also from an economic standpoint. Despite its importance as a reemerging disease, there is no licensed vaccine or specific treatment to prevent CHIK. Many studies have begun to elucidate the pathogenesis of CHIKF and the mechanism of persistent arthralgia, including the role of the adaptive immune response, which is still poorly understood. In addition, the lack of an animal model for chronic infection has limited studies of CHIKV pathogenesis as well as the ability to assess the safety of vaccine candidates currently under development. To address this deficiency, we used recombination activating gene 1 (RAG1-/-) knockout mice, which are deficient in both T and B lymphocytes, to develop a chronic CHIKV infection model. Here, we describe this model as well as its use in evaluating the safety of a live-attenuated vaccine candidate. Chikungunya virus (CHIKV), the etiologic agent of chikungunya fever (CHIK), is a positive sense, single-stranded RNA virus in the genus Alphavirus. Chikungunya fever begins as a flu-like illness, which progresses to severe arthralgia and debilitating arthritis. This syndrome is often self-limited and rarely fatal; however many patients develop persistent arthralgia that can last from months to years. Currently there is no licensed vaccine or specific treatment for CHIKF, leaving current treatment as purely supportive in nature. The role of the adaptive immune system in disease course and viral persistence is still poorly understood. The lack of an animal model of persistent CHIKF has hindered the study of the role of the adaptive immune response and safety testing of vaccine candidates, which are under development. Due to the fact that the vaccine candidate would be deployed in areas where numerous individuals have an impaired adaptive immune system due to malnutrition or disease (HIV/AIDS); it is important to study the safety of the vaccine candidate in immunodeficient animals with no adaptive immune system. In this study we present an animal model of persistent CHIKV in adult mice, which lack an adaptive immune system and demonstrate the safety of a live-attenuated vaccine candidate.
Collapse
|
210
|
Salazar-González JA, Angulo C, Rosales-Mendoza S. Chikungunya virus vaccines: Current strategies and prospects for developing plant-made vaccines. Vaccine 2015; 33:3650-8. [PMID: 26073010 DOI: 10.1016/j.vaccine.2015.05.104] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 05/25/2015] [Accepted: 05/28/2015] [Indexed: 12/18/2022]
Abstract
Chikungunya virus is an emerging pathogen initially found in East Africa and currently spread into the Indian Ocean Islands, many regions of South East Asia, and in the Americas. No licensed vaccines against this eminent pathogen are available and thus intensive research in this field is a priority. This review presents the current scenario on the developments of Chikungunya virus vaccines and identifies the use of genetic engineered plants to develop attractive vaccines. The possible avenues to develop plant-made vaccines with distinct antigenic designs and expression modalities are identified and discussed considering current trends in the field.
Collapse
Affiliation(s)
- Jorge A Salazar-González
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí 78210, SLP, Mexico
| | - Carlos Angulo
- Grupo de Inmunología y Vacunología, Centro de Investigaciones Biológicas del Noroeste, SC., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., C.P. 23096 Mexico City, Mexico
| | - Sergio Rosales-Mendoza
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí 78210, SLP, Mexico.
| |
Collapse
|
211
|
Petitdemange C, Wauquier N, Vieillard V. Control of immunopathology during chikungunya virus infection. J Allergy Clin Immunol 2015; 135:846-855. [PMID: 25843597 DOI: 10.1016/j.jaci.2015.01.039] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 01/20/2015] [Accepted: 01/28/2015] [Indexed: 10/23/2022]
Abstract
After several decades of epidemiologic silence, chikungunya virus (CHIKV) has recently re-emerged, causing explosive outbreaks and reaching the 5 continents. Transmitted through the bite of Aedes species mosquitoes, CHIKV is responsible for an acute febrile illness accompanied by several characteristic symptoms, including cutaneous rash, myalgia, and arthralgia, with the latter sometimes persisting for months or years. Although CHIKV has previously been known as a relatively benign disease, more recent epidemic events have brought waves of increased morbidity and fatality, leading it to become a serious public health problem. The host's immune response plays a crucial role in controlling the infection, but it might also contribute to the promotion of viral spread and immunopathology. This review focuses on the immune responses to CHIKV in human subjects with an emphasis on early antiviral immune responses. We assess recent developments in the understanding of their possible Janus-faced effects in the control of viral infection and pathogenesis. Although preventive vaccination and specific therapies are yet to be developed, exploring this interesting model of virus-host interactions might have a strong effect on the design of novel therapeutic options to minimize immunopathology without impairing beneficial host defenses.
Collapse
Affiliation(s)
| | - Nadia Wauquier
- Sorbonne Universités, UPMC, University of Paris 06, CR7, CIMI-Paris, Paris, France; Metabiota, San Francisco, Calif
| | - Vincent Vieillard
- Sorbonne Universités, UPMC, University of Paris 06, CR7, CIMI-Paris, Paris, France; INSERM, U1135, CIMI-Paris, Paris, France; CNRS, ERL 8255, CIMI-Paris, Paris, France.
| |
Collapse
|
212
|
Chen TH, Jian SW, Wang CY, Lin C, Wang PF, Su CL, Teng HJ, Shu PY, Wu HS. Susceptibility of Aedes albopictus and Aedes aegypti to three imported Chikungunya virus strains, including the E1/226V variant in Taiwan. J Formos Med Assoc 2015; 114:546-52. [DOI: 10.1016/j.jfma.2014.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 12/03/2014] [Accepted: 12/26/2014] [Indexed: 10/24/2022] Open
|
213
|
Goh LYH, Kam YW, Metz SW, Hobson-Peters J, Prow NA, McCarthy S, Smith DW, Pijlman GP, Ng LFP, Hall RA. A sensitive epitope-blocking ELISA for the detection of Chikungunya virus-specific antibodies in patients. J Virol Methods 2015; 222:55-61. [PMID: 26025459 DOI: 10.1016/j.jviromet.2015.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 05/25/2015] [Accepted: 05/25/2015] [Indexed: 01/09/2023]
Abstract
Chikungunya fever (CHIKF) has re-emerged as an arboviral disease that mimics clinical symptoms of other diseases such as dengue, malaria, as well as other alphavirus-related illnesses leading to problems with definitive diagnosis of the infection. Herein we describe the development and evaluation of a sensitive epitope-blocking ELISA (EB-ELISA) capable of specifically detecting anti-chikungunya virus (CHIKV) antibodies in clinical samples. The assay uses a monoclonal antibody (mAb) that binds an epitope on the E2 protein of CHIKV and does not exhibit cross-reactivity to other related alphaviruses. We also demonstrated the use of recombinant CHIK virus-like particles (VLPs) as a safe alternative antigen to infectious virions in the assay. Based on testing of 60 serum samples from patients in the acute or convalescent phase of CHIKV infection, the EB-ELISA provided us with 100% sensitivity, and exhibited 98.5% specificity when Ross River virus (RRV)- or Barmah Forest virus (BFV)-immune serum samples were included. This assay meets the public health demands of a rapid, robust, sensitive and specific, yet simple assay for specifically diagnosing CHIK-infections in humans.
Collapse
Affiliation(s)
- Lucas Y H Goh
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, QLD, Australia
| | - Yiu-Wing Kam
- Laboratory of Microbial Immunity, Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore 138648, Singapore
| | - Stefan W Metz
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Jody Hobson-Peters
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, QLD, Australia
| | - Natalie A Prow
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, QLD, Australia
| | - Suzi McCarthy
- School of Pathology and Laboratory Medicine, The University of Western Australia, Crawley 6009, WA, Australia; Division of Microbiology and Infectious Diseases, PathWest Laboratory Medicine, Nedlands 6009, WA, Australia
| | - David W Smith
- School of Pathology and Laboratory Medicine, The University of Western Australia, Crawley 6009, WA, Australia; Division of Microbiology and Infectious Diseases, PathWest Laboratory Medicine, Nedlands 6009, WA, Australia
| | - Gorben P Pijlman
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Lisa F P Ng
- Laboratory of Microbial Immunity, Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore 138648, Singapore
| | - Roy A Hall
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, QLD, Australia.
| |
Collapse
|
214
|
Abstract
Chikungunya virus (CHIKV) infection is a reemerging pandemic human arboviral disease. CD4+ T cells were previously shown to contribute to joint inflammation in the course of CHIKV infection in mice. The JES6-1 anti-IL-2 antibody selectively expands mouse regulatory T cells (Tregs) by forming a complex with IL-2. In this study, we show that the IL-2 JES6-1-mediated expansion of Tregs ameliorates CHIKV-induced joint pathology. It does so by inhibiting the infiltration of CD4+ T cells due to the induction of anergy in CHIKV-specific CD4+ effector T cells. These findings suggest that activation of Tregs could also become an alternative approach to control CHIKV-mediated disease.
IMPORTANCE Chikungunya virus (CHIKV) has reemerged as a pathogen of global significance. Patients infected with CHIKV suffer from incapacitating joint pain that severely affects their daily functioning. Despite the best efforts, treatment is still inadequate. While T cell-mediated immunopathology in CHIKV infections has been reported, the role of regulatory T cells (Tregs) has not been explored. The JES6-1 anti-interleukin 2 (IL-2) antibody has been demonstrated to selectively expand mouse Tregs by forming a complex with IL-2. We reveal here that IL-2 JES6-1-mediated expansion of Tregs ameliorates CHIKV-induced joint pathology in mice by neutralizing virus-specific CD4+ effector T (Teff) cells. We show that this treatment abrogates the infiltration of pathogenic CD4+ T cells through induction of anergy in CHIKV-specific CD4+ Teff cells. This is the first evidence where the role of Tregs is demonstrated in CHIKV pathogenesis, and its expansion could control virus-mediated immunopathology.
Collapse
|
215
|
Clark DV, Kibuuka H, Millard M, Wakabi S, Lukwago L, Taylor A, Eller MA, Eller LA, Michael NL, Honko AN, Olinger GG, Schoepp RJ, Hepburn MJ, Hensley LE, Robb ML. Long-term sequelae after Ebola virus disease in Bundibugyo, Uganda: a retrospective cohort study. THE LANCET. INFECTIOUS DISEASES 2015; 15:905-12. [PMID: 25910637 DOI: 10.1016/s1473-3099(15)70152-0] [Citation(s) in RCA: 153] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND The limited data available for long-term Ebola virus disease health outcomes suggest that sequelae persist for longer than 1 year after infection. The magnitude of the present outbreak in west Africa necessitates a more complete understanding of the health effects and future medical needs of these patients. METHODS We invited adult survivors of the 2007 Bundibugyo Ebola virus outbreak in Uganda and their contacts to take part in an observational study roughly 29 months after the outbreak. We collected information about health status, functional limitations, and demographics. We collected blood samples for clinical chemistry, haematology, and filovirus antibodies using ELISA. Analyses were restricted to probable and confirmed survivors and their seronegative contacts. FINDINGS We recruited 70 survivors of the 2007 Bundibugyo Ebola virus and 223 contacts. We did analyses for 49 probable and confirmed survivors and 157 seronegative contacts. Survivors of the Bundibugyo Ebola virus were at significantly increased risk of ocular deficits (retro-orbital pain [RR 4·3, 95% CI 1·9-9·6; p<0·0001], blurred vision [1·9, 1·1-3·2; p=0·018]), hearing loss (2·3, 1·2-4·5; p=0·010), difficulty swallowing (2·1, 1·1-3·9; p=0·017), difficulty sleeping (1·9, 1·3-2·8; p=0·001), arthralgias (2·0, 1·1-3·6; p=0·020), and various constitutional symptoms controlling for age and sex. Chronic health problems (prevalence ratio [PR] 2·1, 95% CI 1·2-3·6; p=0·008) and limitations due to memory loss or confusion (PR 5·8, 1·5-22·4; p=0·010) were also reported more frequently by survivors of Bundibugyo Ebola virus. INTERPRETATION Long-term sequelae persist for more than 2 years after Ebola virus disease. Definition of health consequences related to Ebola virus disease could improve patient care for survivors and contribute to understanding of disease pathogenesis. FUNDING Chemical Biological Technologies Directorate, Defense Threat Reduction Agency.
Collapse
Affiliation(s)
- Danielle V Clark
- Walter Reed Army Institute of Research, Silver Spring, MD, USA; Naval Medical Research Center, Biological Defense Research Directorate, Fort Detrick, MD, USA.
| | - Hannah Kibuuka
- Makerere University Walter Reed Project, Kampala, Uganda
| | - Monica Millard
- Makerere University Walter Reed Project, Kampala, Uganda
| | - Salim Wakabi
- Makerere University Walter Reed Project, Kampala, Uganda
| | | | - Alison Taylor
- Makerere University Walter Reed Project, Kampala, Uganda
| | | | | | - Nelson L Michael
- Walter Reed Army Institute of Research, Silver Spring, MD, USA; US Military HIV Research Program, Bethesda, MD, USA
| | - Anna N Honko
- Virology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA; Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, MD, USA
| | - Gene G Olinger
- Virology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA; Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, MD, USA
| | - Randal J Schoepp
- Diagnostic Systems Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Matthew J Hepburn
- Medical Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Lisa E Hensley
- Virology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA; Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, MD, USA
| | | |
Collapse
|
216
|
Ekchariyawat P, Hamel R, Bernard E, Wichit S, Surasombatpattana P, Talignani L, Thomas F, Choumet V, Yssel H, Desprès P, Briant L, Missé D. Inflammasome signaling pathways exert antiviral effect against Chikungunya virus in human dermal fibroblasts. INFECTION GENETICS AND EVOLUTION 2015; 32:401-8. [PMID: 25847693 DOI: 10.1016/j.meegid.2015.03.025] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 03/20/2015] [Accepted: 03/21/2015] [Indexed: 12/20/2022]
Abstract
Arboviruses represent an emerging threat to human. They are transmitted to vertebrates by the bite of infected arthropods. Early transmission to vertebrates is initiated by skin puncture and deposition of virus in this organ. However, events at the bite site remain largely unknown. Here, we report that Chikungunya virus (CHIKV) and West Nile virus (WNV), despite belonging to distinct viral families, elicit a common antiviral signature in primary human dermal fibroblasts, attesting for the up regulation of interferon signaling pathways and leading to an increased expression of IFN-β, interleukins and chemokines. Remarkably, CHIKV and WNV enhance IL-1β expression and induce maturation of caspase-1, indicating the capacity of these pathogens to elicit activation of the inflammasome program in resident skin cells. CHIKV and WNV also induce the expression of the inflammasome sensor AIM2 in dermal fibroblasts, whereas inhibition of caspase-1 and AIM2 with siRNA interferes with both CHIKV- and WNV-induced IL-1β production by these cells. Finally, inhibition of the inflammasome via caspase-1 silencing was found to enhance CHIKV replication in dermal fibroblasts. Together, these results indicate that the skin contributes to the pro-inflammatory and anti-viral microenvironment via the activation of the inflammasome in the early stages following infection with arboviruses.
Collapse
Affiliation(s)
| | - Rodolphe Hamel
- Laboratoire MIVEGEC, UMR 224 IRD/CNRS/UM1, Montpellier, France
| | - Eric Bernard
- Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé, CNRS-UMR 5236/UM1/UM2, Montpellier, France
| | | | | | - Loïc Talignani
- Laboratoire MIVEGEC, UMR 224 IRD/CNRS/UM1, Montpellier, France
| | - Frédéric Thomas
- Laboratoire MIVEGEC, UMR 224 IRD/CNRS/UM1, Montpellier, France
| | - Valérie Choumet
- Unit Environment and Infectious Risks, Institut Pasteur, Paris, France
| | - Hans Yssel
- Centre d'Immunologie et des Maladies Infectieuses, Inserm, U1135, Sorbonne Universités, UPMC, APHP Hôpital Pitié-Salpêtrière, Paris, France
| | - Philippe Desprès
- Département infections et Epidémiologie, Institut Pasteur, 75724 Paris et UMR PIMIT (I2T team), Université de La Réunion, INSERM U1187, CNRS 9192, IRD 249, GIP-CYROI, 97491 Saint Clotilde, La Réunion, France
| | - Laurence Briant
- Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé, CNRS-UMR 5236/UM1/UM2, Montpellier, France
| | - Dorothée Missé
- Laboratoire MIVEGEC, UMR 224 IRD/CNRS/UM1, Montpellier, France.
| |
Collapse
|
217
|
Foo SS, Chen W, Taylor A, Sheng KC, Yu X, Teng TS, Reading PC, Blanchard H, Garlanda C, Mantovani A, Ng LFP, Herrero LJ, Mahalingam S. Role of pentraxin 3 in shaping arthritogenic alphaviral disease: from enhanced viral replication to immunomodulation. PLoS Pathog 2015; 11:e1004649. [PMID: 25695775 PMCID: PMC4335073 DOI: 10.1371/journal.ppat.1004649] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 01/01/2015] [Indexed: 11/21/2022] Open
Abstract
The rising prevalence of arthritogenic alphavirus infections, including chikungunya virus (CHIKV) and Ross River virus (RRV), and the lack of antiviral treatments highlight the potential threat of a global alphavirus pandemic. The immune responses underlying alphavirus virulence remain enigmatic. We found that pentraxin 3 (PTX3) was highly expressed in CHIKV and RRV patients during acute disease. Overt expression of PTX3 in CHIKV patients was associated with increased viral load and disease severity. PTX3-deficient (PTX3(-/-)) mice acutely infected with RRV exhibited delayed disease progression and rapid recovery through diminished inflammatory responses and viral replication. Furthermore, binding of the N-terminal domain of PTX3 to RRV facilitated viral entry and replication. Thus, our study demonstrates the pivotal role of PTX3 in shaping alphavirus-triggered immunity and disease and provides new insights into alphavirus pathogenesis.
Collapse
Affiliation(s)
- Suan-Sin Foo
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Weiqiang Chen
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Adam Taylor
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Kuo-Ching Sheng
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Xing Yu
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Terk-Shin Teng
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Patrick C. Reading
- WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Helen Blanchard
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | - Cecilia Garlanda
- Humanitas Clinical and Research Center, Department of Inflammation and Immunology, Rozzano, Italy
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Department of Inflammation and Immunology, Rozzano, Italy
- Department of Biotechnology and Translational Medicine, University of Milan, Milano, Italy
| | - Lisa F. P. Ng
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lara J. Herrero
- Institute for Glycomics, Griffith University, Gold Coast, Australia
| | | |
Collapse
|
218
|
Teng TS, Kam YW, Lee B, Hapuarachchi HC, Wimal A, Ng LC, Ng LFP. A Systematic Meta-analysis of Immune Signatures in Patients With Acute Chikungunya Virus Infection. J Infect Dis 2015; 211:1925-35. [PMID: 25635123 PMCID: PMC4442625 DOI: 10.1093/infdis/jiv049] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 01/20/2015] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Individuals infected with chikungunya virus (CHIKV) normally exhibit a variety of clinical manifestations during the acute phase of infection. However, studies in different patient cohorts have revealed that disease manifestations vary in frequency. METHODS Disease profiles between patients with acute CHIKV-infection and febrile patients without CHIKV were compared and examined to determine whether any clinical presentations were associated with the clinical outcome of CHIKV infection. Circulatory immune mediators profiles were then characterized and compared with data from 14 independent patient cohort studies. The particular immune mediator signature that defines acute CHIKV infection was determined. RESULTS Our findings revealed a specific pattern of clinical presentations of joint-specific arthralgia from this CHIKV cohort. More importantly, we identified an immune mediator signature dominated by proinflammatory cytokines, which include interferon α and γ and interleukin 2, 2R, 6, 7, 12, 15, 17, and 18, across different patient cohorts of CHIKV load associated with arthralgia. CONCLUSIONS To our knowledge, this is the first study that associated levels of CHIKV load with arthralgia as an indicator of acute CHIKV infection. Importantly, our findings also revealed specific immune mediator signatures that can be used to better define CHIKV infection.
Collapse
Affiliation(s)
- Terk-Shin Teng
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis
| | - Yiu-Wing Kam
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis
| | - Bernett Lee
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis
| | | | - Abeyewickreme Wimal
- Department of Parasitology, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - Lee-Ching Ng
- Environmental Health Institute, National Environment Agency, Singapore
| | - Lisa F P Ng
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis Institute of Infection and Global Health, University of Liverpool, United Kingdom
| |
Collapse
|
219
|
|
220
|
Moyen N, Thiberville SD, Pastorino B, Nougairede A, Thirion L, Mombouli JV, Dimi Y, Leparc-Goffart I, Capobianchi MR, Lepfoundzou AD, de Lamballerie X. First reported chikungunya fever outbreak in the republic of Congo, 2011. PLoS One 2014; 9:e115938. [PMID: 25541718 PMCID: PMC4277398 DOI: 10.1371/journal.pone.0115938] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 11/29/2014] [Indexed: 12/21/2022] Open
Abstract
Background Chikungunya is an Aedes -borne disease characterised by febrile arthralgia and responsible for massive outbreaks. We present a prospective clinical cohort study and a retrospective serological study relating to a CHIK outbreak, in the Republic of Congo in 2011. Methodology and Findings We analysed 317 suspected cases, of which 308 (97.2%) lived in the city of Brazzaville (66.6% in the South area). Amongst them, 37 (11.7%) were CHIKV+ve patients (i.e., biologically confirmed by a real-time RT-PCR assay), of whom 36 (97.3%) had fever, 22 (66.7%) myalgia and 32 (86.5%) arthralgia. All tested negative for dengue. The distribution of incident cases within Brazzaville districts was compared with CHIKV seroprevalence before the outbreak (34.4% in 517 blood donors), providing evidence for previous circulation of CHIKV. We applied a CHIK clinical score to 126 patients recruited within the two first day of illness (including 28 CHIKV+ves (22.2%)) with sensitivity (78.6%) and specificity (72.4%) values comparing with those of the referent study in Reunion Island. The negative predictive value was high (92%), but the positive predictive value (45%) indicate poor potential contribution to medical practice to identify CHIKV+ve patients in low prevalence outbreaks. However, the score allowed a slightly more accurate follow-up of the evolution of the outbreak than the criterion "fever+arthralgia". The complete sequencing of a Congolase isolate (Brazza_MRS1) demonstrated belonging to the East/Central/South African lineage and was further used for producing a robust genome-scale CHIKV phylogenetic analysis. Conclusions/Significance We describe the first Chikungunya outbreak declared in the Republic of Congo. The seroprevalence study conducted amongst blood donors before outbreak provided evidence for previous CHIKV circulation. We suggest that a more systematic survey of the entomological situation and of arbovirus circulation is necessary in Central Africa for better understanding the environmental, microbiological and sociological determinants of emergence.
Collapse
Affiliation(s)
- Nanikaly Moyen
- Aix Marseille University, IRD French Institute of Research for Development, EHESP French School of Public Health, EPV UMR_D 190 "Emergence des Pathologies Virales" 13005, Marseille, France
- * E-mail:
| | - Simon-Djamel Thiberville
- Aix Marseille University, IRD French Institute of Research for Development, EHESP French School of Public Health, EPV UMR_D 190 "Emergence des Pathologies Virales" 13005, Marseille, France
| | - Boris Pastorino
- Aix Marseille University, IRD French Institute of Research for Development, EHESP French School of Public Health, EPV UMR_D 190 "Emergence des Pathologies Virales" 13005, Marseille, France
| | - Antoine Nougairede
- Aix Marseille University, IRD French Institute of Research for Development, EHESP French School of Public Health, EPV UMR_D 190 "Emergence des Pathologies Virales" 13005, Marseille, France
| | - Laurence Thirion
- Aix Marseille University, IRD French Institute of Research for Development, EHESP French School of Public Health, EPV UMR_D 190 "Emergence des Pathologies Virales" 13005, Marseille, France
| | | | - Yannick Dimi
- Centre National de Transfusion Sanguine, Brazzaville, Republic of Congo
| | - Isabelle Leparc-Goffart
- French National Reference Centre for Arboviruses, IRBA Armed Forces Biomedical Research Institute, 13013, Marseille, France
| | - Maria Rosaria Capobianchi
- Laboratory of Virology, National Institute for Infectious Diseases "L. Spallanzani", Via Portuense 292, 00149, Rome, Italy
| | | | - Xavier de Lamballerie
- Aix Marseille University, IRD French Institute of Research for Development, EHESP French School of Public Health, EPV UMR_D 190 "Emergence des Pathologies Virales" 13005, Marseille, France
| |
Collapse
|
221
|
Chang LJ, Dowd KA, Mendoza FH, Saunders JG, Sitar S, Plummer SH, Yamshchikov G, Sarwar UN, Hu Z, Enama ME, Bailer RT, Koup RA, Schwartz RM, Akahata W, Nabel GJ, Mascola JR, Pierson TC, Graham BS, Ledgerwood JE. Safety and tolerability of chikungunya virus-like particle vaccine in healthy adults: a phase 1 dose-escalation trial. Lancet 2014; 384:2046-52. [PMID: 25132507 DOI: 10.1016/s0140-6736(14)61185-5] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND Chikungunya virus--a mosquito-borne alphavirus--is endemic in Africa and south and southeast Asia and has recently emerged in the Caribbean. No drugs or vaccines are available for treatment or prevention. We aimed to assess the safety, tolerability, and immunogenicity of a new candidate vaccine. METHODS VRC 311 was a phase 1, dose-escalation, open-label clinical trial of a virus-like particle (VLP) chikungunya virus vaccine, VRC-CHKVLP059-00-VP, in healthy adults aged 18-50 years who were enrolled at the National Institutes of Health Clinical Center (Bethesda, MD, USA). Participants were assigned to sequential dose level groups to receive vaccinations at 10 μg, 20 μg, or 40 μg on weeks 0, 4, and 20, with follow-up for 44 weeks after enrolment. The primary endpoints were safety and tolerability of the vaccine. Secondary endpoints were chikungunya virus-specific immune responses assessed by ELISA and neutralising antibody assays. This trial is registered with ClinicalTrials.gov, NCT01489358. FINDINGS 25 participants were enrolled from Dec 12, 2011, to March 22, 2012, into the three dosage groups: 10 μg (n=5), 20 μg (n=10), and 40 μg (n=10). The protocol was completed by all five participants at the 10 μg dose, all ten participants at the 20 μg dose, and eight of ten participants at the 40 μg dose; non-completions were for personal circumstances unrelated to adverse events. 73 vaccinations were administered. All injections were well tolerated, with no serious adverse events reported. Neutralising antibodies were detected in all dose groups after the second vaccination (geometric mean titres of the half maximum inhibitory concentration: 2688 in the 10 μg group, 1775 in the 20 μg group, and 7246 in the 40 μg group), and a significant boost occurred after the third vaccination in all dose groups (10 μg group p=0·0197, 20 μg group p<0·0001, and 40 μg group p<0·0001). 4 weeks after the third vaccination, the geometric mean titres of the half maximum inhibitory concentration were 8745 for the 10 μg group, 4525 for the 20 μg group, and 5390 for the 40 μg group. INTERPRETATION The chikungunya VLP vaccine was immunogenic, safe, and well tolerated. This study represents an important step in vaccine development to combat this rapidly emerging pathogen. Further studies should be done in a larger number of participants and in more diverse populations. FUNDING Intramural Research Program of the Vaccine Research Center, National Institute of Allergy and Infectious Diseases, and National Institutes of Health.
Collapse
Affiliation(s)
- Lee-Jah Chang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kimberly A Dowd
- Viral Pathogenesis Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Floreliz H Mendoza
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jamie G Saunders
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sandra Sitar
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sarah H Plummer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Galina Yamshchikov
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Uzma N Sarwar
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Zonghui Hu
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mary E Enama
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert T Bailer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Richard M Schwartz
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Wataru Akahata
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Gary J Nabel
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Theodore C Pierson
- Viral Pathogenesis Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julie E Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
222
|
Chen W, Foo SS, Li RW, Smith PN, Mahalingam S. Osteoblasts from osteoarthritis patients show enhanced susceptibility to Ross River virus infection associated with delayed type I interferon responses. Virol J 2014; 11:189. [PMID: 25407789 PMCID: PMC4252017 DOI: 10.1186/s12985-014-0189-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/26/2014] [Indexed: 11/29/2022] Open
Abstract
Background Arthritogenic alphaviruses such as Ross River virus (RRV) and chikungunya virus (CHIKV) have caused widespread outbreaks of chronic polyarthritis. The inflammatory responses in alphavirus-induced arthritis and osteoarthritis (OA) share many similar features, which suggests the possibility of exacerbated alphavirus-induced bone pathology in individuals with pre-existing OA. Here, we investigated the susceptibility of osteoblasts (OBs) from OA patients to RRV infection and dissected the immune mechanisms elicited from infection. Methods Primary hOBs obtained from trabecular bone of healthy donors and OA patients were infected with RRV. Infectivity and viral replication were determined using flow cytometry and plaque assay, respectively. Real-time PCR was performed to determine expression kinetics of type I interferon (IFN)-related immune mediators and osteotropic factors. Results OA hOBs showed enhanced RRV infectivity and replication during infection, which was associated with delayed induction of IFN-β and RIG-I expression. Enhanced susceptibility of OA hOBs to RRV was associated with a more pronounced increase in RANKL/OPG ratio and expression of osteotropic factors (IL-6, IL-1β, TNF-α and CCL2) in comparison to RRV-infected healthy hOBs. Conclusions Delayed activation of type I IFN-signalling pathway may have contributed to enhanced susceptibility to RRV infection in hOBs from OA patients. RRV-induced increases in RANKL/OPG ratio and expression of osteotropic factors that favour bone resorption, which may be exacerbated during osteoarthritis. This study provides the novel insight that osteoarthritis may be a risk factor for exacerbated arthritogenic alphaviral infection.
Collapse
Affiliation(s)
- Weiqiang Chen
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, 4222, Australia.
| | - Suan-Sin Foo
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, 4222, Australia.
| | - Rachel W Li
- Trauma and Orthopaedic Research Unit Laboratory, The Medical School, The Australian National University, Garran Rd, Canberra, ACT 2601, Australia.
| | - Paul N Smith
- Department of Orthopaedic Surgery, Trauma and Orthopaedic Research Unit, The Canberra Hospital, Canberra, ACT 2605, Australia.
| | - Suresh Mahalingam
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, 4222, Australia.
| |
Collapse
|
223
|
Neutralization activity of patient sera collected during the 2008-2009 Chikungunya outbreak in Thailand. J Clin Microbiol 2014; 53:184-90. [PMID: 25378567 DOI: 10.1128/jcm.02744-14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Chikungunya virus (CHIKV) infection typically causes fever, rash, myalgia, and arthralgia and sometimes results in recurrent joint pain or, in severe cases, neurological disorders or death. How CHIKV infection leads to prolonged or severe symptoms is still not well understood. In this study, we examined the neutralization (NT) titer of 98 serum samples collected from patients during the 2008-2009 chikungunya outbreak in Thailand. While all serum samples showed neutralizing activity, virus was detected in 58% of the serum samples. When we analyzed a possible association between virus and antibody titers and the presence of typical symptoms of CHIKV infection, fever and joint pain, there was no significant association except that the number of patients with fever was over three times more than the number of those without fever when CHIKV was detectable in serum. This study indicates that although neutralizing antibody is critical to eliminate CHIKV, it appears not to be the main factor associated with clinical symptoms in some cases, so that other aspects of immune responses, such as those involving proinflammatory mediators and adaptive immune cells, should be considered altogether.
Collapse
|
224
|
Arthritogenic alphaviruses: new insights into arthritis and bone pathology. Trends Microbiol 2014; 23:35-43. [PMID: 25449049 DOI: 10.1016/j.tim.2014.09.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 09/18/2014] [Accepted: 09/19/2014] [Indexed: 01/01/2023]
Abstract
Arthritogenic alphaviral infection begins as a febrile illness and often progresses to joint pain and rheumatic symptoms that are described as polyarthritis. Alphaviral arthritis and classical arthritides share many similar cellular and immune mediators involved in their pathogenesis. Recent in vitro and in vivo evidence suggests that bone loss resulting from increased expression of bone resorption mediators may accompany alphaviral infection. In addition, several longitudinal studies have reported more severe and delayed recovery of alphaviral disease in patients with pre-existing arthritic conditions. This review aims to provide insights into alphavirus-induced bone loss and focuses on aspects of disease exacerbation in patients with underlying arthritis and on possible therapeutic targets.
Collapse
|
225
|
Venugopalan A, Ghorpade RP, Chopra A. Cytokines in acute chikungunya. PLoS One 2014; 9:e111305. [PMID: 25343623 PMCID: PMC4208842 DOI: 10.1371/journal.pone.0111305] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 09/30/2014] [Indexed: 01/08/2023] Open
Abstract
Introduction Acute chikungunya (CHIKV) is predominantly an acute onset of excruciatingly painful, self-limiting musculoskeletal (MSK) arbovirus illness and this was further reported by us during the 2006 Indian epidemic [Chopra et al. Epidemiol Infect 2012]. Selected serum cytokines profile in subjects within one month of onset of illness is being presented. Methods Out of 509 clinical CHIKV cases (43% population) identified during a rural population survey, 225 subjects consented blood investigations. 132 examined within 30 days of febrile onset are the study cohort. Anti-CHIKV IgM and IgG antibodies tested by immunochromatography and indirect immunofluorescence respectively. Interferons (IFN)-α, -β and -γ, Interferon Gamma-Induced Protein-10 (CXCL-10/IP-10), Tumor Necrosis Factor-α (TNF-α), Interleukin-1β (IL-1β), Interleukin-6 (IL-6), Interleukin-13 (IL-13), Monocyte Chemoattractant Protein-1 (MCP-1), Interleukin–4 (IL-4) and Interleukin–10 (IL-10) performed by ELISA. Samples collected from neighboring community a year prior to the epidemic used as healthy controls. Results Seropositivity for anti-CHIKV IgM and IgG was 65% and 52% respectively. IFN-α, IFN-β, IFN-γ, CXCL10/IP-10 and IL-1β showed intense response in early acute phase. Cytokines (particularly TNF-α, MCP-1, IL-4, IL-6 and IL-10) was maximum in extended symptomatic phase and remained elevated in recovered subjects. Higher (p<0.05) IFN and IL-4 seen in patients seropositive for anti-CHIKV IgG. Elderly cases (≥65 years) showed elevated cytokines (except IFN) and anti-CHIKV antibodies near similar to younger subjects. Significant correlations (p<0.05) found between cytokines and clinical features (fatigue, low back ache, myalgia) and anti-CHIKV antibodies. Conclusion An intense cytokine milieu was evident in the early and immediate persistent symptomatic phase and in recovered subjects. Early persistent IgM and lower IgG to anti-CHKV and intense Th2 cytokine phenotype seem to be associated with delay in resolution of MSK symptoms. Intriguingly, maximum TNF-α, IL-6 and IL-13 with low anti-CHIKV IgM response found in subjects recovered from CHIKV within one month of illness.
Collapse
|
226
|
Bindarit, an inhibitor of monocyte chemotactic protein synthesis, protects against bone loss induced by chikungunya virus infection. J Virol 2014; 89:581-93. [PMID: 25339772 DOI: 10.1128/jvi.02034-14] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED The recent global resurgence of arthritogenic alphaviruses, in particular chikungunya virus (CHIKV), highlights an urgent need for the development of therapeutic intervention strategies. While there has been significant progress in defining the pathophysiology of alphaviral disease, relatively little is known about the mechanisms involved in CHIKV-induced arthritis or potential therapeutic options to treat the severe arthritic symptoms associated with infection. Here, we used microcomputed tomographic (μCT) and histomorphometric analyses to provide previously undescribed evidence of reduced bone volume in the proximal tibial epiphysis of CHIKV-infected mice compared to the results for mock controls. This was associated with a significant increase in the receptor activator of nuclear factor-κB ligand/osteoprotegerin (RANKL/OPG) ratio in infected murine joints and in the serum of CHIKV patients. The expression levels of the monocyte chemoattractant proteins (MCPs), including MCP-1/CCL2, MCP-2/CCL8, and MCP-3/CCL7, were also highly elevated in joints of CHIKV-infected mice, accompanied by increased cellularity within the bone marrow in tibial epiphysis and ankle joints. Both this effect and CHIKV-induced bone loss were significantly reduced by treatment with the MCP inhibitor bindarit. Collectively, these findings demonstrate a unique role for MCPs in promoting CHIKV-induced osteoclastogenesis and bone loss during disease and suggest that inhibition of MCPs with bindarit may be an effective therapy for patients affected with alphavirus-induced bone loss. IMPORTANCE Arthritogenic alphaviruses, including chikungunya virus (CHIKV) and Ross River virus (RRV), cause worldwide outbreaks of polyarthritis, which can persist in patients for months following infection. Previous studies have shown that host proinflammatory soluble factors are associated with CHIKV disease severity. Furthermore, it is established that chemokine (C-C motif) ligand 2 (CCL2/MCP-1) is important in cellular recruitment and inducing bone-resorbing osteoclast (OC) formation. Here, we show that CHIKV replicates in bone and triggers bone loss by increasing the RANKL/OPG ratio. CHIKV infection results in MCP-induced cellular infiltration in the inflamed joints, and bone loss can be ameliorated by treatment with an MCP-inhibiting drug, bindarit. Taken together, our data reveal a previously undescribed role for MCPs in CHIKV-induced bone loss: one of recruiting monocytes/OC precursors to joint sites and thereby favoring a pro-osteoclastic microenvironment. This suggests that bindarit may be an effective treatment for alphavirus-induced bone loss and arthritis in humans.
Collapse
|
227
|
van den Doel P, Volz A, Roose JM, Sewbalaksing VD, Pijlman GP, van Middelkoop I, Duiverman V, van de Wetering E, Sutter G, Osterhaus ADME, Martina BEE. Recombinant modified vaccinia virus Ankara expressing glycoprotein E2 of Chikungunya virus protects AG129 mice against lethal challenge. PLoS Negl Trop Dis 2014; 8:e3101. [PMID: 25188230 PMCID: PMC4154657 DOI: 10.1371/journal.pntd.0003101] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 07/07/2014] [Indexed: 01/04/2023] Open
Abstract
Chikungunya virus (CHIKV) infection is characterized by rash, acute high fever, chills, headache, nausea, photophobia, vomiting, and severe polyarthralgia. There is evidence that arthralgia can persist for years and result in long-term discomfort. Neurologic disease with fatal outcome has been documented, although at low incidences. The CHIKV RNA genome encodes five structural proteins (C, E1, E2, E3 and 6K). The E1 spike protein drives the fusion process within the cytoplasm, while the E2 protein is believed to interact with cellular receptors and therefore most probably constitutes the target of neutralizing antibodies. We have constructed recombinant Modified Vaccinia Ankara (MVA) expressing E3E2, 6KE1, or the entire CHIKV envelope polyprotein cassette E3E26KE1. MVA is an appropriate platform because of its demonstrated clinical safety and its suitability for expression of various heterologous proteins. After completing the immunization scheme, animals were challenged with CHIV-S27. Immunization of AG129 mice with MVAs expressing E2 or E3E26KE1 elicited neutralizing antibodies in all animals and provided 100% protection against lethal disease. In contrast, 75% of the animals immunized with 6KE1 were protected against lethal infection. In conclusion, MVA expressing the glycoprotein E2 of CHIKV represents as an immunogenic and effective candidate vaccine against CHIKV infections.
Collapse
Affiliation(s)
- Petra van den Doel
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Asisa Volz
- Institute for Infectious Diseases and Zoonoses, University of Munich LMU, Munich, Germany
| | - Jouke M. Roose
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Gorben P. Pijlman
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
| | | | - Vincent Duiverman
- Erasmus Medical Center Laboratory Animal Science Center (EDC), Rotterdam, The Netherlands
| | - Eva van de Wetering
- Erasmus Medical Center Laboratory Animal Science Center (EDC), Rotterdam, The Netherlands
| | - Gerd Sutter
- Institute for Infectious Diseases and Zoonoses, University of Munich LMU, Munich, Germany
| | - Albert D. M. E. Osterhaus
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
- Artemis One Health, Utrecht, The Netherlands
| | - Byron E. E. Martina
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
- Artemis One Health, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
228
|
Horcada ML, Díaz-Calderón C, Garrido L. Chikungunya fever. Rheumatic manifestations of an emerging disease in Europe. ACTA ACUST UNITED AC 2014; 11:161-4. [PMID: 25192946 DOI: 10.1016/j.reuma.2014.07.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 06/23/2014] [Accepted: 07/11/2014] [Indexed: 10/24/2022]
Abstract
Chikungunya fever is a viral disease caused by an alphavirus belonging to the Togaviridae family, transmitted by several species of Aedes mosquitoes: Aedes aegypti and Aedes albopictus (A. albopictus). It is endemic in Africa and Asia with recurrent outbreaks. It is an emerging disease and cases in Europe transmitted by A. albopictus have been established in Mediterranean areas. The first autochthonous cases detected on the Caribbean islands suppose a serious threat of spreading disease to America, which so far has been disease free. Clinical symptoms begin abruptly with fever, skin rash and polyarthritis. Although mortality is low, a high percentage of patients develop a chronic phase defined by persistent arthritis for months or even years. A severe immune response is responsible for joint inflammation. The absence of specific treatment and lack of vaccine requires detailed studies about its immunopathogenesis in order to determine the most appropriate target.
Collapse
Affiliation(s)
- M Loreto Horcada
- Servicio de Reumatología, Complejo Hospitalario de Navarra, Pamplona, Navarra, España.
| | | | - Laura Garrido
- Servicio de Reumatología, Complejo Hospitalario de Navarra, Pamplona, Navarra, España
| |
Collapse
|
229
|
|
230
|
Goody MF, Sullivan C, Kim CH. Studying the immune response to human viral infections using zebrafish. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 46:84-95. [PMID: 24718256 PMCID: PMC4067600 DOI: 10.1016/j.dci.2014.03.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 03/31/2014] [Accepted: 03/31/2014] [Indexed: 05/24/2023]
Abstract
Humans and viruses have a long co-evolutionary history. Viral illnesses have and will continue to shape human history: from smallpox, to influenza, to HIV, and beyond. Animal models of human viral illnesses are needed in order to generate safe and effective antiviral medicines, adjuvant therapies, and vaccines. These animal models must support the replication of human viruses, recapitulate aspects of human viral illnesses, and respond with conserved immune signaling cascades. The zebrafish is perhaps the simplest, most commonly used laboratory model organism in which innate and/or adaptive immunity can be studied. Herein, we will discuss the current zebrafish models of human viral illnesses and the insights they have provided. We will highlight advantages of early life stage zebrafish and the importance of innate immunity in human viral illnesses. We will also discuss viral characteristics to consider before infecting zebrafish with human viruses as well as predict other human viruses that may be able to infect zebrafish.
Collapse
Affiliation(s)
- Michelle F Goody
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA
| | - Con Sullivan
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA
| | - Carol H Kim
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME 04469, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA.
| |
Collapse
|
231
|
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that causes acute fever and acute and chronic musculoskeletal pain in humans. Since 2004, CHIKV has caused millions of cases of disease in the Indian Ocean region and has emerged in new areas, including Europe, the Middle East, and the Pacific region. The mosquito vectors for this virus are globally distributed in tropical and temperate zones, providing the opportunity for CHIKV to continue to expand into new geographic regions. In October 2013, locally acquired cases of CHIKV infection were identified on the Caribbean island of Saint Martin, signaling the arrival of the virus in the Western Hemisphere. In just 9 months, CHIKV has spread to 22 countries in the Caribbean and Central and South America, resulting in hundreds of thousands of cases. CHIKV disease can be highly debilitating, and large epidemics have severe economic consequences. Thus, there is an urgent need for continued research into the epidemiology, pathogenesis, prevention, and treatment of these infections.
Collapse
|
232
|
Smith DR. Global protein profiling studies of chikungunya virus infection identify different proteins but common biological processes. Rev Med Virol 2014; 25:3-18. [PMID: 25066270 DOI: 10.1002/rmv.1802] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 06/26/2014] [Accepted: 06/26/2014] [Indexed: 12/24/2022]
Abstract
Chikungunya fever (CHIKF) caused by the mosquito-transmitted chikungunya virus (CHIKV) swept into international prominence from late 2005 as an epidemic of CHIKF spread around countries surrounding the Indian Ocean. Although significant advances have been made in understanding the pathobiology of CHIKF, numerous questions still remain. In the absence of commercially available specific drugs to treat the disease, or a vaccine to prevent the diseases, the questions have particular significance. A number of studies have used global proteome analysis to increase our understanding of the process of CHIKV infection using a number of different experimental techniques and experimental systems. In all, over 700 proteins have been identified in nine different analyses by five different groups as being differentially regulated. Remarkably, only a single protein, eukaryotic elongation factor 2, has been identified by more than two different groups as being differentially regulated during CHIKV infection. This review provides a critical overview of the studies that have used global protein profiling to understand CHIKV infection and shows that while a broad consensus is emerging on which biological processes are altered during CHIKV infection, this consensus is poorly supported in terms of consistent identification of any key proteins mediating those biological processes.
Collapse
Affiliation(s)
- Duncan R Smith
- Institute of Molecular Biosciences, Mahidol University, Bangkok, Thailand; Center for Emerging and Neglected Infectious Diseases, Mahidol University, Bangkok, Thailand
| |
Collapse
|
233
|
Yaseen HM, Simon F, Deparis X, Marimoutou C. Identification of initial severity determinants to predict arthritis after chikungunya infection in a cohort of French gendarmes. BMC Musculoskelet Disord 2014; 15:249. [PMID: 25059583 PMCID: PMC4121301 DOI: 10.1186/1471-2474-15-249] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 06/06/2014] [Indexed: 11/27/2022] Open
Abstract
Background The objective was to identify severity characteristics of initial chikungunya infection (CHIKV) stages associated with post-CHIKV arthritis and arthralgia. Methods French gendarmes exposed to the 2005–2006 CHIKV epidemic in Reunion Island who completed the 2006 (self-reporting acute and early chronic [median: 6 months] symptoms) and 2008 (Endpoint [median: 30 months]: self-perceived recovery and rheumatic disorders (RDs)) surveys were included. Multinomial logistic regression and multiple correspondence analysis (MCA) were used. Arthralgia was defined by joint pain and/or stiffness and arthritis by joint swelling in addition to pain and/or stiffness. Results In 2008, 124 (31.3%)/403 participants (101 CHIKV+/302 CHIKV-) reported arthralgia and 57 (14.1%) arthritis. The multivariate model kept CHIKV infection, comorbidity and acute stage depressed mood as independent prognostic factors for both arthralgia and arthritis, but found early chronic stage RD as the main determinant of the same RD two years later. The MCA performed with the 85 CHIKV + patients who answered the question on self-perceived recovery enabled the calculation of severity scores based on initial symptoms that were strongly associated with persistent arthritis and, to a lesser extent, to arthralgia in bivariate analyses. The MCA graph clearly distinguished arthritis as the only RD associated with early severity indicators represented by sick leave, joint swelling and depressed mood during the acute stage, and early chronification of arthritis and depressed mood. Conclusion Initial CHIKV severity predicted recovery, with higher severity associated with arthritis and lower severity with arthralgia. More interestingly, specific markers of post-CHIKV arthritis, which can easily be used by clinicians for case management, were identified.
Collapse
Affiliation(s)
| | | | | | - Catherine Marimoutou
- Centre d'Epidémiologie et Santé Publique des Armées (CESPA), BP 40026, Marseille 13568 cedex 02, France.
| |
Collapse
|
234
|
Hawman DW, Burrack KS, Morrison TE. Mechanisms underlying the pathogenesis of arthritogenic alphaviruses: host immune responses and virus persistence. Future Virol 2014; 9:441-444. [PMID: 33907560 DOI: 10.2217/fvl.14.27] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- David W Hawman
- Department of Microbiology, University of Colorado School of Medicine, 12800 East 19th Avenue, Mail Stop 8333, Aurora, CO 80045, USA
| | - Kristina S Burrack
- Department of Microbiology, University of Colorado School of Medicine, 12800 East 19th Avenue, Mail Stop 8333, Aurora, CO 80045, USA
| | - Thomas E Morrison
- Department of Microbiology, University of Colorado School of Medicine, 12800 East 19th Avenue, Mail Stop 8333, Aurora, CO 80045, USA
| |
Collapse
|
235
|
Chikungunya viruses that escape monoclonal antibody therapy are clinically attenuated, stable, and not purified in mosquitoes. J Virol 2014; 88:8213-26. [PMID: 24829346 DOI: 10.1128/jvi.01032-14] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Chikungunya virus (CHIKV) is a reemerging mosquito-transmitted alphavirus that causes epidemics of debilitating polyarthritis in humans. A prior study identified two anti-CHIKV monoclonal antibodies ([MAbs] CHK-152 and CHK-166) against the E2 and E1 structural proteins, which had therapeutic efficacy in immunocompetent and immunocompromised mice. Combination MAb therapy was required as administration of a single MAb resulted in the rapid selection of neutralization escape variants and treatment failure in mice. Here, we initially evaluated the efficacy of combination MAb therapy in a nonhuman primate model of CHIKV infection. Treatment of rhesus macaques with CHK-152 and CHK-166 reduced viral spread and infection in distant tissue sites and also neutralized reservoirs of infectious virus. Escape viruses were not detected in the residual viral RNA present in tissues and organs of rhesus macaques. To evaluate the possible significance of MAb resistance, we engineered neutralization escape variant viruses (E1-K61T, E2-D59N, and the double mutant E1-K61T E2-D59N) that conferred resistance to CHK-152 and CHK-166 and tested them for fitness in mosquito cells, mammalian cells, mice, and Aedes albopictus mosquitoes. In both cell culture and mosquitoes, the mutant viruses grew equivalently and did not revert to wild-type (WT) sequence. All escape variants showed evidence of mild clinical attenuation, with decreased musculoskeletal disease at early times after infection in WT mice and a prolonged survival time in immunocompromised Ifnar1(-/-) mice. Unexpectedly, this was not associated with decreased infectivity, and consensus sequencing from tissues revealed no evidence of reversion or compensatory mutations. Competition studies with CHIKV WT also revealed no fitness compromise of the double mutant (E1-K61T E2-D59N) neutralization escape variant in WT mice. Collectively, our study suggests that neutralization escape viruses selected during combination MAb therapy with CHK-152 plus CHK-166 retain fitness, cause less severe clinical disease, and likely would not be purified during the enzootic cycle. IMPORTANCE Chikungunya virus (CHIKV) causes explosive epidemics of acute and chronic arthritis in humans in Africa, the Indian subcontinent, and Southeast Asia and recently has spread to the New World. As there are no approved vaccines or therapies for human use, the possibility of CHIKV-induced debilitating disease is high in many parts of the world. To this end, our laboratory recently generated a combination monoclonal antibody therapy that aborted lethal and arthritogenic disease in wild-type and immunocompromised mice when administered as a single dose several days after infection. In this study, we show the efficacy of the antibody combination in nonhuman primates and also evaluate the significance of possible neutralization escape mutations in mosquito and mammalian cells, mice, and Aedes albopictus vector mosquitoes. Our experiments show that escape viruses from combination antibody therapy cause less severe CHIKV clinical disease, retain fitness, and likely would not be purified by mosquito vectors.
Collapse
|
236
|
Arthritogenic alphaviral infection perturbs osteoblast function and triggers pathologic bone loss. Proc Natl Acad Sci U S A 2014; 111:6040-5. [PMID: 24733914 DOI: 10.1073/pnas.1318859111] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Arthritogenic alphaviruses including Ross River virus (RRV), Sindbis virus, and chikungunya virus cause worldwide outbreaks of musculoskeletal disease. The ability of alphaviruses to induce bone pathologies remains poorly defined. Here we show that primary human osteoblasts (hOBs) can be productively infected by RRV. RRV-infected hOBs produced high levels of inflammatory cytokine including IL-6. The RANKL/OPG ratio was disrupted in the synovial fluid of RRV patients, and this was accompanied by an increase in serum Tartrate-resistant acid phosphatase 5b (TRAP5b) levels. Infection of bone cells with RRV was validated using an established RRV murine model. In wild-type mice, infectious virus was detected in the femur, tibia, patella, and foot, together with reduced bone volume in the tibial epiphysis and vertebrae detected by microcomputed tomographic (µCT) analysis. The RANKL/OPG ratio was also disrupted in mice infected with RRV; both this effect and the bone loss were blocked by treatment with an IL-6 neutralizing antibody. Collectively, these findings provide previously unidentified evidence that alphavirus infection induces bone loss and that OBs are capable of producing proinflammatory mediators during alphavirus-induced arthralgia. The perturbed RANKL/OPG ratio in RRV-infected OBs may therefore contribute to bone loss in alphavirus infection.
Collapse
|
237
|
Bourjot M, Leyssen P, Neyts J, Dumontet V, Litaudon M. Trigocherrierin A, a potent inhibitor of chikungunya virus replication. Molecules 2014; 19:3617-27. [PMID: 24662077 PMCID: PMC6270878 DOI: 10.3390/molecules19033617] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 03/12/2014] [Accepted: 03/17/2014] [Indexed: 11/29/2022] Open
Abstract
Trigocherrierin A (1) and trigocherriolide E (2), two new daphnane diterpenoid orthoesters (DDOs), and six chlorinated analogues, trigocherrins A, B, F and trigocherriolides A–C, were isolated from the leaves of Trigonostemon cherrieri. Their structures were identified by mass spectrometry, extensive one- and two-dimensional NMR spectroscopy and through comparison with data reported in the literature. These compounds are potent and selective inhibitors of chikungunya virus (CHIKV) replication. Among the DDOs isolated, compound 1 exhibited the strongest anti-CHIKV activity (EC50 = 0.6 ± 0.1 µM, SI = 71.7).
Collapse
Affiliation(s)
- Mélanie Bourjot
- EA4267 Epithelial Functions and Dysfunctions, UFR of Medical and Pharmaceutical Sciences, 19 rue Ambroise Paré, 25030 Besançon, France.
| | - Pieter Leyssen
- Rega Institute for Medical Research (KU Leuven), Minderbroedersstraat 10, B3000, Leuven, Belgium.
| | - Johan Neyts
- Rega Institute for Medical Research (KU Leuven), Minderbroedersstraat 10, B3000, Leuven, Belgium.
| | - Vincent Dumontet
- Gif Research Center, Institute of Chemistry of Natural Substances (ICSN), CNRS, Labex CEBA, 1, avenue de la Terrasse, 91198 Gif sur Yvette Cedex, France.
| | - Marc Litaudon
- Gif Research Center, Institute of Chemistry of Natural Substances (ICSN), CNRS, Labex CEBA, 1, avenue de la Terrasse, 91198 Gif sur Yvette Cedex, France.
| |
Collapse
|
238
|
Gardner CL, Hritz J, Sun C, Vanlandingham DL, Song TY, Ghedin E, Higgs S, Klimstra WB, Ryman KD. Deliberate attenuation of chikungunya virus by adaptation to heparan sulfate-dependent infectivity: a model for rational arboviral vaccine design. PLoS Negl Trop Dis 2014; 8:e2719. [PMID: 24587470 PMCID: PMC3930508 DOI: 10.1371/journal.pntd.0002719] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 01/13/2014] [Indexed: 12/16/2022] Open
Abstract
Mosquito-borne chikungunya virus (CHIKV) is a positive-sense, single-stranded RNA virus from the genus Alphavirus, family Togaviridae, which causes fever, rash and severe persistent polyarthralgia in humans. Since there are currently no FDA licensed vaccines or antiviral therapies for CHIKV, the development of vaccine candidates is of critical importance. Historically, live-attenuated vaccines (LAVs) for protection against arthropod-borne viruses have been created by blind cell culture passage leading to attenuation of disease, while maintaining immunogenicity. Attenuation may occur via multiple mechanisms. However, all examined arbovirus LAVs have in common the acquisition of positively charged amino acid substitutions in cell-surface attachment proteins that render virus infection partially dependent upon heparan sulfate (HS), a ubiquitously expressed sulfated polysaccharide, and appear to attenuate by retarding dissemination of virus particles in vivo. We previously reported that, like other wild-type Old World alphaviruses, CHIKV strain, La Réunion, (CHIKV-LR), does not depend upon HS for infectivity. To deliberately identify CHIKV attachment protein mutations that could be combined with other attenuating processes in a LAV candidate, we passaged CHIKV-LR on evolutionarily divergent cell-types. A panel of single amino acid substitutions was identified in the E2 glycoprotein of passaged virus populations that were predicted to increase electrostatic potential. Each of these substitutions was made in the CHIKV-LR cDNA clone and comparisons of the mutant viruses revealed surface exposure of the mutated residue on the spike and sensitivity to competition with the HS analog, heparin, to be primary correlates of attenuation in vivo. Furthermore, we have identified a mutation at E2 position 79 as a promising candidate for inclusion in a CHIKV LAV. With the adaptation of chikungunya virus (CHIKV) to transmission by the Aedes albopictus mosquito, a pandemic has occurred resulting in four to six million human infections, and the virus continues to become endemic in new regions, most recently in the Caribbean. CHIKV can cause debilitating polyarthralgia, lasting for weeks to years, and there are currently no licensed vaccines or antiviral therapies available. While an investigational live-attenuated vaccine (LAV) exists, problems with reactogenicity have precluded its licensure. The purpose of the current study was to: i) devise an in vitro passage procedure that reliably generates a panel of CHIKV envelope glycoprotein mutations for screening as vaccine candidates; ii) determine the position of the mutations in the three-dimensional structure of the alphavirus spike complex and their effect on electrostatic potential; iii) determine the attenuation characteristics of each mutation in a murine model of CHIKV musculoskeletal disease; and iv) to identify in vitro assays examining the dependency of infection upon HS that correlate with attenuation and localization in the glycoprotein spike. This approach provides a paradigm for the rational design of future LAVs for CHIKV and other mosquito-borne viruses, by deliberately selecting and combining attenuating processes.
Collapse
Affiliation(s)
- Christina L. Gardner
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology & Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jozef Hritz
- CEITEC, Masaryk University, Brno, Czech Republic
| | - Chengqun Sun
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology & Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Dana L. Vanlandingham
- Department of Diagnostic Medicine & Pathobiology, Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, United States of America
| | - Timothy Y. Song
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Elodie Ghedin
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Stephen Higgs
- Department of Diagnostic Medicine & Pathobiology, Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, United States of America
| | - William B. Klimstra
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology & Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Kate D. Ryman
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology & Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
239
|
Reddy V, Mani RS, Desai A, Ravi V. Correlation of plasma viral loads and presence of Chikungunya IgM antibodies with cytokine/chemokine levels during acute Chikungunya virus infection. J Med Virol 2014; 86:1393-401. [DOI: 10.1002/jmv.23875] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2013] [Indexed: 01/01/2023]
Affiliation(s)
- Vijayalakshmi Reddy
- Department of Neurovirology; National Institute of Mental Health and Neurosciences; Bangalore Karnataka India
| | - Reeta Subramaniam Mani
- Department of Neurovirology; National Institute of Mental Health and Neurosciences; Bangalore Karnataka India
| | - Anita Desai
- Department of Neurovirology; National Institute of Mental Health and Neurosciences; Bangalore Karnataka India
| | - Vasanthapuram Ravi
- Department of Neurovirology; National Institute of Mental Health and Neurosciences; Bangalore Karnataka India
| |
Collapse
|
240
|
A novel poxvirus-based vaccine, MVA-CHIKV, is highly immunogenic and protects mice against chikungunya infection. J Virol 2014; 88:3527-47. [PMID: 24403588 DOI: 10.1128/jvi.03418-13] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
UNLABELLED There is a need to develop a single and highly effective vaccine against the emerging chikungunya virus (CHIKV), which causes a severe disease in humans. Here, we have generated and characterized the immunogenicity profile and the efficacy of a novel CHIKV vaccine candidate based on the highly attenuated poxvirus vector modified vaccinia virus Ankara (MVA) expressing the CHIKV C, E3, E2, 6K, and E1 structural genes (termed MVA-CHIKV). MVA-CHIKV was stable in cell culture, expressed the CHIKV structural proteins, and triggered the cytoplasmic accumulation of Golgi apparatus-derived membranes in infected human cells. Furthermore, MVA-CHIKV elicited robust innate immune responses in human macrophages and monocyte-derived dendritic cells, with production of beta interferon (IFN-β), proinflammatory cytokines, and chemokines. After immunization of C57BL/6 mice with a homologous protocol (MVA-CHIKV/MVA-CHIKV), strong, broad, polyfunctional, and durable CHIKV-specific CD8(+) T cell responses were elicited. The CHIKV-specific CD8(+) T cells were preferentially directed against E1 and E2 proteins and, to a lesser extent, against C protein. CHIKV-specific CD8(+) memory T cells of a mainly effector memory phenotype were also induced. The humoral arm of the immune system was significantly induced, as MVA-CHIKV elicited high titers of neutralizing antibodies against CHIKV. Remarkably, a single dose of MVA-CHIKV protected all mice after a high-dose challenge with CHIKV. In summary, MVA-CHIKV is an effective vaccine against chikungunya virus infection that induced strong, broad, highly polyfunctional, and long-lasting CHIKV-specific CD8(+) T cell responses, together with neutralizing antibodies against CHIKV. These results support the consideration of MVA-CHIKV as a potential vaccine candidate against CHIKV. IMPORTANCE We have developed a novel vaccine candidate against chikungunya virus (CHIKV) based on the highly attenuated poxvirus vector modified vaccinia virus Ankara (MVA) expressing the CHIKV C, E3, E2, 6K, and E1 structural genes (termed MVA-CHIKV). Our findings revealed that MVA-CHIKV is a highly effective vaccine against chikungunya virus, with a single dose of the vaccine protecting all mice after a high-dose challenge with CHIKV. Furthermore, MVA-CHIKV is highly immunogenic, inducing strong innate responses: high, broad, polyfunctional, and long-lasting CHIKV-specific CD8(+) T cell responses, together with neutralizing antibodies against CHIKV. This work provides a potential vaccine candidate against CHIKV.
Collapse
|
241
|
Kashyap RS, Morey S, Bhullar S, Baheti N, Chandak N, Purohit H, Taori G, Daginawala H. Determination of Toll-like receptor-induced cytokine profiles in the blood and cerebrospinal fluid of Chikungunya patients. Neuroimmunomodulation 2014; 21:338-46. [PMID: 24776821 DOI: 10.1159/000358240] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 12/23/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Chikungunya infection caused by Chikungunya virus (CHIKV) is an inflammatory disease affecting the joints and may also lead to neurological complications. We investigated a panel of human Toll-like receptor (TLR)-induced cytokines in Chikungunya patients with and without neurological complications. METHODS In a case-control study, a panel of 12 cytokines and chemokines, TNF-α, IFN-α, IL-1β, IL-6, IL-12, IL-17A, IL-8, monocyte chemotactic protein (MCP)-1, RANTES, interferon (IFN)-γ-induced protein (IP)-10, monokine induced by IFN-γ (MIG) and thymus and activation-regulated chemokine (TARC), was analysed using a conventional ELISA protocol in the serum samples of Chikungunya patients without neurological complications and in the cerebrospinal fluid (CSF) and paired serum samples of Chikungunya patients with neurological complications. RESULTS The levels of 3 cytokines, IL-1β, IL-17A and IL-8, and 4 chemokines, MCP-1, RANTES, IP-10 and TARC, were raised in serum samples of Chikungunya patients without neurological complications, whereas, 4 cytokines, TNF-α, IFN-α, IL-6 and IL-8, and 4 chemokines, MCP-1, RANTES, MIG and TARC, were elevated in CSF samples of Chikungunya patients with neurological complications. Moreover, the levels of IL-6 and IL-8 cytokines were significantly elevated in the CSF compared to paired serum samples in Chikungunya patients with neurological complications. CONCLUSIONS In CHIKV infection, multiple cytokines are elevated in serum and CSF. The elevation in IL-6 and IL-8 cytokines in CSF correlates with neurological involvement.
Collapse
Affiliation(s)
- Rajpal Singh Kashyap
- Biochemistry Research Laboratory, Central India Institute of Medical Sciences, Nagpur, India
| | | | | | | | | | | | | | | |
Collapse
|
242
|
Puttamallesh VN, Sreenivasamurthy SK, Singh PK, Harsha HC, Ganjiwale A, Broor S, Pandey A, Narayana J, Prasad TSK. Proteomic profiling of serum samples from chikungunya-infected patients provides insights into host response. Clin Proteomics 2013; 10:14. [PMID: 24124767 PMCID: PMC3879382 DOI: 10.1186/1559-0275-10-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 09/17/2013] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Chikungunya is a highly debilitating febrile illness caused by Chikungunya virus, a single-stranded RNA virus, which is transmitted by Aedes aegypti or Aedes albopictus mosquito species. The pathogenesis and host responses in individuals infected with the chikungunya virus are not well understood at the molecular level. We carried out proteomic profiling of serum samples from chikungunya patients in order to identify molecules associated with the host response to infection by this virus. RESULTS Proteomic profiling of serum obtained from the infected individuals resulted in identification of 569 proteins. Of these, 63 proteins were found to be differentially expressed (≥ 2-fold) in patient as compared to control sera. These differentially expressed proteins were involved in various processes such as lipid metabolism, immune response, transport, signal transduction and apoptosis. CONCLUSIONS This is the first report providing a global proteomic profile of serum samples from individuals infected with the chikungunya virus. Our data provide an insight into the proteins that are involved as host response factors during an infection. These proteins include clusterin, apolipoproteins and S100A family of proteins.
Collapse
Affiliation(s)
- Vinuth N Puttamallesh
- Institute of Bioinformatics, International Technology Park, Bangalore 560 066, India
| | | | - Pradeep Kumar Singh
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi 110 029, India
| | - H C Harsha
- Institute of Bioinformatics, International Technology Park, Bangalore 560 066, India
| | - Anjali Ganjiwale
- Microtest Innovations Pvt. Limited, International Technology Park, Bangalore 560 066, India
| | - Shobha Broor
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi 110 029, India
| | - Akhilesh Pandey
- Institute of Bioinformatics, International Technology Park, Bangalore 560 066, India
- McKusick-Nathans Institute of Genetic Medicine and Departments of Biological Chemistry, Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore 21205 MD, USA
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore 21205 MD, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore 21205 MD, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore 21205 MD, USA
| | - Jayasuryan Narayana
- Microtest Innovations Pvt. Limited, International Technology Park, Bangalore 560 066, India
| | - T S Keshava Prasad
- Institute of Bioinformatics, International Technology Park, Bangalore 560 066, India
| |
Collapse
|
243
|
Chiam CW, Chan YF, Loong SK, Yong SSJ, Hooi PS, Sam IC. Real-time polymerase chain reaction for diagnosis and quantitation of negative strand of chikungunya virus. Diagn Microbiol Infect Dis 2013; 77:133-7. [DOI: 10.1016/j.diagmicrobio.2013.06.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 06/10/2013] [Accepted: 06/13/2013] [Indexed: 12/23/2022]
|
244
|
Herrero LJ, Sheng KC, Jian P, Taylor A, Her Z, Herring BL, Chow A, Leo YS, Hickey MJ, Morand EF, Ng LF, Bucala R, Mahalingam S. Macrophage migration inhibitory factor receptor CD74 mediates alphavirus-induced arthritis and myositis in murine models of alphavirus infection. ARTHRITIS AND RHEUMATISM 2013; 65:2724-36. [PMID: 23896945 PMCID: PMC3796577 DOI: 10.1002/art.38090] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 07/09/2013] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Arthrogenic alphaviruses such as Ross River virus (RRV) and chikungunya virus (CHIKV) circulate worldwide. This virus class causes debilitating illnesses that are characterized by arthritis, arthralgia, and myalgia. In previous studies, we identified macrophage migration inhibitory factor (MIF) as a critical inflammatory factor in the pathogenesis of alphaviral diseases. The present study was undertaken to characterize the role of CD74, a cell surface receptor of MIF, in both RRV- and CHIKV-induced alphavirus arthritides. METHODS Mouse models of RRV and CHIKV infection were used to investigate the immunopathogenesis of arthritic alphavirus infection. The role of CD74 was assessed using histologic analysis, real-time polymerase chain reaction, flow cytometry, and plaque assay. RESULTS In comparison to wild-type mice, CD74-/- mice developed only mild clinical features and had low levels of tissue damage. Leukocyte infiltration, characterized predominantly by inflammatory monocytes and natural killer cells, was substantially reduced in the infected tissue of CD74-/- mice, but production of proinflammatory cytokines and chemokines was not decreased. CD74 deficiency was associated with increased monocyte apoptosis, but had no effect on monocyte migratory capacity. Consistent with these findings, alphaviral infection resulted in a dose-dependent up-regulation of CD74 expression in human peripheral blood mononuclear cells, and serum MIF levels were significantly elevated in patients with RRV or CHIKV infection. CONCLUSION CD74 appears to regulate immune responses to alphaviral infection through its effects on cellular recruitment and survival. These findings suggest that both MIF and CD74 play a critical role in mediating alphaviral disease, and blocking these factors with novel therapeutic agents could substantially ameliorate the pathologic manifestations.
Collapse
MESH Headings
- Alphavirus Infections/complications
- Alphavirus Infections/pathology
- Animals
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigens, Differentiation, B-Lymphocyte/physiology
- Apoptosis/physiology
- Arthritis, Infectious/etiology
- Arthritis, Infectious/pathology
- Arthritis, Infectious/physiopathology
- Cells, Cultured
- Chemokines/metabolism
- Chikungunya virus/physiology
- Cytokines/metabolism
- Disease Models, Animal
- Female
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/physiology
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Monocytes/pathology
- Myositis/pathology
- Myositis/physiopathology
- Myositis/virology
- Receptors, Immunologic/deficiency
- Receptors, Immunologic/genetics
- Receptors, Immunologic/physiology
- Ross River virus/physiology
- Severity of Illness Index
Collapse
Affiliation(s)
- Lara J. Herrero
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, Australia
| | - Kuo-Ching Sheng
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, Australia
| | - Peng Jian
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, Australia
| | - Adam Taylor
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, Australia
| | - Zhisheng Her
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Belinda L. Herring
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, Australia
| | - Angela Chow
- Communicable Disease Centre, Tan Tock Seng Hospital, Singapore
| | - Yee-Sin Leo
- Communicable Disease Centre, Tan Tock Seng Hospital, Singapore
| | | | - Eric F. Morand
- Centre for Inflammatory Diseases, Monash University, VIC, Australia
| | - Lisa F.P. Ng
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511 USA
| | - Suresh Mahalingam
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, Australia
| |
Collapse
|
245
|
Phuklia W, Kasisith J, Modhiran N, Rodpai E, Thannagith M, Thongsakulprasert T, Smith DR, Ubol S. Osteoclastogenesis induced by CHIKV-infected fibroblast-like synoviocytes: a possible interplay between synoviocytes and monocytes/macrophages in CHIKV-induced arthralgia/arthritis. Virus Res 2013; 177:179-88. [PMID: 24012515 DOI: 10.1016/j.virusres.2013.08.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 08/26/2013] [Accepted: 08/27/2013] [Indexed: 02/03/2023]
Abstract
Fibroblast-like synoviocytes are known to migrate from joint to joint and are proposed to be one of the key players in the inflammatory cascade amplification in rheumatoid arthritis patients. In the recent CHIKV epidemic, patients developed arthritis-like syndrome and the synoviocyte is one of the suspected players in CHIKV-induced polyarthritis. Thus, to learn more on this syndrome, the responses of fibroblast-like synoviocytes to chikungunya virus (CHIKV) infection, and the interaction between CHIKV-infected synoviocytes and phagocytes, were investigated. Primary human fibroblast-like synoviocyte (HFLS) cultures were infected with clinical isolates of CHIKV at an MOI of 0.001pfu/cell. Data indicated that HFLS are permissive to CHIKV replication, generating peak titers of 10(5)-10(6)pfu/ml. Interestingly, CHIKV-infected HFLS cultures secreted mainly the mediators that are responsible for phagocytes recruitment and differentiation (RANKL, IL-6, IL-8 and MCP-1) but not arthritogenic mediators (TNF-α, IL-1β, MMP-1, MMP-2 or MMP-13). The interaction between CHIKV-infected synoviocytes and phagocytes was studied using UV-irradiated, CHIKV-infected HFLS supernatant. Data revealed that supernatants from CHIKV-infected HFLS cultures not only induced migration of primary human monocytes, but also drove monocytes/macrophages into osteoclast-like cells. These differentiated osteoclast-like cells produced high levels of TNF-α and IL-6, principal mediators of arthritis. This data suggests a potential interplay between infected HFLS and recruiting phagocytes which may responsible for the arthralgia/arthritis in CHIKV-infected patients.
Collapse
Affiliation(s)
- Weerawat Phuklia
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | | | | | | | | | | | | |
Collapse
|
246
|
Palha N, Guivel-Benhassine F, Briolat V, Lutfalla G, Sourisseau M, Ellett F, Wang CH, Lieschke GJ, Herbomel P, Schwartz O, Levraud JP. Real-time whole-body visualization of Chikungunya Virus infection and host interferon response in zebrafish. PLoS Pathog 2013; 9:e1003619. [PMID: 24039582 PMCID: PMC3764224 DOI: 10.1371/journal.ppat.1003619] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 07/29/2013] [Indexed: 12/23/2022] Open
Abstract
Chikungunya Virus (CHIKV), a re-emerging arbovirus that may cause severe disease, constitutes an important public health problem. Herein we describe a novel CHIKV infection model in zebrafish, where viral spread was live-imaged in the whole body up to cellular resolution. Infected cells emerged in various organs in one principal wave with a median appearance time of ∼14 hours post infection. Timing of infected cell death was organ dependent, leading to a shift of CHIKV localization towards the brain. As in mammals, CHIKV infection triggered a strong type-I interferon (IFN) response, critical for survival. IFN was mainly expressed by neutrophils and hepatocytes. Cell type specific ablation experiments further demonstrated that neutrophils play a crucial, unexpected role in CHIKV containment. Altogether, our results show that the zebrafish represents a novel valuable model to dynamically visualize replication, pathogenesis and host responses to a human virus. Chikungunya, a re-emerging disease caused by a mosquito-transmitted virus, is an important public health problem. We developed a zebrafish model for chikungunya virus infection. For the first time, rise and death of virus-infected cells could be live imaged in the entire body of a vertebrate. We observed a widespread wave of apparition of newly infected cells during the first day after inoculation of the virus. We then found that infected cells died at a strongly organ-dependent rate, accounting for the progressive shift of virus localization. Notably, the virus persisted in the brain despite apparent recovery of infected zebrafish. We found this recovery to be critically dependent on the host type I interferon response. Surprisingly, we identified neutrophils as a major cell population expressing interferon and controlling chikungunya virus.
Collapse
Affiliation(s)
- Nuno Palha
- Institut Pasteur, Macrophages et Développement de l'Immunité, Department of Developmental and Stem Cells Biology, Paris, France
- CNRS URA2578, Paris, France
- Université Pierre et Marie Curie, Paris, France
| | | | - Valérie Briolat
- Institut Pasteur, Macrophages et Développement de l'Immunité, Department of Developmental and Stem Cells Biology, Paris, France
- CNRS URA2578, Paris, France
| | - Georges Lutfalla
- CNRS UMR5235, Dynamiques des Interactions Membranaires et Pathologiques, Montpellier, France
- Université Montpellier II, Montpellier, France
| | - Marion Sourisseau
- Institut Pasteur, Virus et Immunité, Department of Virology, Paris, France
- CNRS URA3015, Paris, France
| | - Felix Ellett
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Chieh-Huei Wang
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Graham J. Lieschke
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Philippe Herbomel
- Institut Pasteur, Macrophages et Développement de l'Immunité, Department of Developmental and Stem Cells Biology, Paris, France
- CNRS URA2578, Paris, France
| | - Olivier Schwartz
- Institut Pasteur, Virus et Immunité, Department of Virology, Paris, France
- CNRS URA3015, Paris, France
| | - Jean-Pierre Levraud
- Institut Pasteur, Macrophages et Développement de l'Immunité, Department of Developmental and Stem Cells Biology, Paris, France
- CNRS URA2578, Paris, France
- * E-mail:
| |
Collapse
|
247
|
Molecular mechanisms involved in the pathogenesis of alphavirus-induced arthritis. BIOMED RESEARCH INTERNATIONAL 2013; 2013:973516. [PMID: 24069610 PMCID: PMC3771267 DOI: 10.1155/2013/973516] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Accepted: 07/22/2013] [Indexed: 11/17/2022]
Abstract
Arthritogenic alphaviruses, including Ross River virus (RRV), Chikungunya virus (CHIKV), Sindbis virus (SINV), Mayaro virus (MAYV), O'nyong-nyong virus (ONNV), and Barmah Forest virus (BFV), cause incapacitating and long lasting articular disease/myalgia. Outbreaks of viral arthritis and the global distribution of these diseases point to the emergence of arthritogenic alphaviruses as an important public health problem. This review discusses the molecular mechanisms involved in alphavirus-induced arthritis, exploring the recent data obtained with in vitro systems and in vivo studies using animal models and samples from patients. The factors associated to the extension and persistence of symptoms are highlighted, focusing on (a) virus replication in target cells, and tissues, including macrophages and muscle cells; (b) the inflammatory and immune responses with recruitment and activation of macrophage, NK cells and T lymphocytes to the lesion focus and the increase of inflammatory mediators levels; and (c) the persistence of virus or viral products in joint and muscle tissues. We also discuss the importance of the establishment of novel animal models to test new molecular targets and to develop more efficient and selective drugs to treat these diseases.
Collapse
|
248
|
Theilacker C, Held J, Allering L, Emmerich P, Schmidt-Chanasit J, Kern WV, Panning M. Prolonged polyarthralgia in a German traveller with Mayaro virus infection without inflammatory correlates. BMC Infect Dis 2013; 13:369. [PMID: 23927600 PMCID: PMC3750572 DOI: 10.1186/1471-2334-13-369] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 08/06/2013] [Indexed: 01/18/2023] Open
Abstract
Background Mayaro virus is endemic in South America and sporadic outbreaks have been described. It causes a dengue-like febrile illness accompanied by severe and long-lasting polyarthralgias. Outside endemic regions, however, the disease is not well known and can be misdiagnosed as dengue. International travellers are at risk to acquire Mayaro virus and due to increased worldwide travel infectious disease specialists need to be aware of such rare clinical entities. Case presentation We report the first Mayaro virus infection imported into Germany. A 20-year-old woman developed fever, myalgia, maculopapular rash, and polyarthralgias following a 10-day trip in the Rurrenabaque region of Bolivia. Severe polyarthralgias persisted for 5 months and were treated with non-steroidal anti-inflammatory drugs. Serological analysis demonstrated Mayaro virus-specific-IgM and -IgG antibodies two months after onset of symptoms. Except for CXCL8/IL-8 other proinflammatory chemokines and cytokines were unremarkable at this time. Conclusions Dissemination of knowledge on rare disease might improve patient management. Understanding the inherent features of Mayaro virus infection and how the virus interacts with its host are essential for optimal patient care and therapy.
Collapse
|
249
|
Chirathaworn C, Poovorawan Y, Lertmaharit S, Wuttirattanakowit N. Cytokine levels in patients with chikungunya virus infection. ASIAN PAC J TROP MED 2013; 6:631-634. [PMID: 23790334 DOI: 10.1016/s1995-7645(13)60108-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 11/15/2012] [Accepted: 12/15/2012] [Indexed: 10/26/2022] Open
Abstract
OBJECTIVE To investigate cytokine profile in patients with chikungunya virus (CHIKV) infection. METHODS Twenty eight pairs of serum samples collected from CHIKV infected patients during the outbreak of chikungunya fever in South Thailand in 2008 were obtained. A multiple cytokine assay for detection of 17 cytokines was performed. RESULTS In the acute stage of CHIKV infection, the patients had significantly higher levels of interleukin-6, granulocyte colony-stimulating factor, granulocyte-macrophage colony-stimulating factor, monocyte chemotactic protein 1 and tumor necrosis factor alpha than the control (P<0.001, P=0.023, P=0.015, P<0.001 and P=0.024, respectively). When the disease developed to the recovery stage, the patients had significantly lower levels of interleukin-6, granulocyte-macrophage colony-stimulating factor, monocyte chemotactic protein 1 and macrophage inflammatory protein beta than in the acute stage (P<0.001). CONCLUSIONS This study provides additional information that these cytokines could play roles in pathogenesis of CHIKV infection and could be used as disease biomarkers or drug targets.
Collapse
Affiliation(s)
- Chintana Chirathaworn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| | | | | | | |
Collapse
|
250
|
Messaoudi I, Vomaske J, Totonchy T, Kreklywich CN, Haberthur K, Springgay L, Brien JD, Diamond MS, DeFilippis VR, Streblow DN. Chikungunya virus infection results in higher and persistent viral replication in aged rhesus macaques due to defects in anti-viral immunity. PLoS Negl Trop Dis 2013; 7:e2343. [PMID: 23936572 PMCID: PMC3723534 DOI: 10.1371/journal.pntd.0002343] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 06/17/2013] [Indexed: 01/06/2023] Open
Abstract
Chikungunya virus (CHIKV) is a re-emerging mosquito-borne Alphavirus that causes a clinical disease involving fever, myalgia, nausea and rash. The distinguishing feature of CHIKV infection is the severe debilitating poly-arthralgia that may persist for several months after viral clearance. Since its re-emergence in 2004, CHIKV has spread from the Indian Ocean region to new locations including metropolitan Europe, Japan, and even the United States. The risk of importing CHIKV to new areas of the world is increasing due to high levels of viremia in infected individuals as well as the recent adaptation of the virus to the mosquito species Aedes albopictus. CHIKV re-emergence is also associated with new clinical complications including severe morbidity and, for the first time, mortality. In this study, we characterized disease progression and host immune responses in adult and aged Rhesus macaques infected with either the recent CHIKV outbreak strain La Reunion (LR) or the West African strain 37997. Our results indicate that following intravenous infection and regardless of the virus used, Rhesus macaques become viremic between days 1–5 post infection. While adult animals are able to control viral infection, aged animals show persistent virus in the spleen. Virus-specific T cell responses in the aged animals were reduced compared to adult animals and the B cell responses were also delayed and reduced in aged animals. Interestingly, regardless of age, T cell and antibody responses were more robust in animals infected with LR compared to 37997 CHIKV strain. Taken together these data suggest that the reduced immune responses in the aged animals promotes long-term virus persistence in CHIKV-LR infected Rhesus monkeys. Chikungunya virus (CHIKV) is a re-emerging Alphavirus that has caused recent massive outbreaks in the Indian Ocean region. In addition, outbreaks have been documented in Europe and elsewhere in the world, initiated by infected travelers returning to their homelands. The recent outbreak strains possess extended vector range and as such, raise the potential of CHIKV outbreaks in the Southeastern parts of the United States. In this study, we examined CHIKV immunity in adult and aged Rhesus macaques following infection with two different CHIKV strains (recent outbreak strain CHIKV-LR and a West African Strain CHIKV-37997). CHIKV-LR causes persistent infection in the aged animals and replicates, on average, to higher levels than CHIKV-37997. Irrespective of the viral strain used, aged animals had delayed and/or reduced immunity compared to adult animals. Our data support the clinical findings of CHIKV susceptibility in vulnerable populations including the aged and provide mechanistic evidence that an effective immune response directed against the virus is required for preventing persistent CHIKV infection.
Collapse
Affiliation(s)
- Ilhem Messaoudi
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
- * E-mail: (IM); (DNS)
| | - Jennifer Vomaske
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Thomas Totonchy
- Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Craig N. Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Kristen Haberthur
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Laura Springgay
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - James D. Brien
- Departments of Molecular Microbiology, Medicine, Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Michael S. Diamond
- Departments of Molecular Microbiology, Medicine, Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Victor R. DeFilippis
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Daniel N. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
- * E-mail: (IM); (DNS)
| |
Collapse
|