301
|
Abstract
In this chapter, we describe 73 zoonotic viruses that were isolated in Northern Eurasia and that belong to the different families of viruses with a single-stranded RNA (ssRNA) genome. The family includes viruses with a segmented negative-sense ssRNA genome (families Bunyaviridae and Orthomyxoviridae) and viruses with a positive-sense ssRNA genome (families Togaviridae and Flaviviridae). Among them are viruses associated with sporadic cases or outbreaks of human disease, such as hemorrhagic fever with renal syndrome (viruses of the genus Hantavirus), Crimean–Congo hemorrhagic fever (CCHFV, Nairovirus), California encephalitis (INKV, TAHV, and KHATV; Orthobunyavirus), sandfly fever (SFCV and SFNV, Phlebovirus), Tick-borne encephalitis (TBEV, Flavivirus), Omsk hemorrhagic fever (OHFV, Flavivirus), West Nile fever (WNV, Flavivirus), Sindbis fever (SINV, Alphavirus) Chikungunya fever (CHIKV, Alphavirus) and others. Other viruses described in the chapter can cause epizootics in wild or domestic animals: Geta virus (GETV, Alphavirus), Influenza A virus (Influenzavirus A), Bhanja virus (BHAV, Phlebovirus) and more. The chapter also discusses both ecological peculiarities that promote the circulation of these viruses in natural foci and factors influencing the occurrence of epidemic and epizootic outbreaks
Collapse
|
302
|
Mutations conferring a noncytotoxic phenotype on chikungunya virus replicons compromise enzymatic properties of nonstructural protein 2. J Virol 2014; 89:3145-62. [PMID: 25552719 DOI: 10.1128/jvi.03213-14] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Chikungunya virus (CHIKV) (genus Alphavirus) has a positive-sense RNA genome. CHIKV nonstructural protein 2 (nsP2) proteolytically processes the viral nonstructural polyprotein, possesses nucleoside triphosphatase (NTPase), RNA triphosphatase, and RNA helicase activities, and induces cytopathic effects in vertebrate cells. Although alphaviral nsP2 mutations can result in a noncytotoxic phenotype, the effects of such mutations on nsP2 enzymatic activities are not well understood. In this study, we introduced a P718G (PG) mutation and selected for additional mutations in CHIKV nsP2 that resulted in a CHIKV replicon with a noncytotoxic phenotype in BHK-21 cells. Combinations of PG and either an E117K (EK) substitution or a GEEGS sequence insertion after residue T647 (5A) markedly reduced RNA synthesis; however, neither PG nor 5A prevented nsP2 nuclear translocation. Introducing PG into recombinant nsP2 inhibited proteolytic cleavage of nsP1/nsP2 and nsP3/nsP4 sites, reduced GTPase and RNA helicase activities, and abolished RNA stimulation of GTPase activity. 5A and EK modulated the effects of PG. However, only the RNA helicase activity of nsP2 was reduced by both of these mutations, suggesting that defects in this activity may be linked to a noncytotoxic phenotype. These results increase our understanding of the molecular basis for the cytotoxicity that accompanies alphaviral replication. Furthermore, adaptation of the CHIKV replicon containing both 5A and PG allowed the selection of a CHIKV replicon with adaptive mutations in nsP1 and nsP3 that enable persistence in human cell line. Such cell lines represent valuable experimental systems for discovering host factors and for screening inhibitors of CHIKV replication at lower biosafety levels. IMPORTANCE CHIKV is a medically important pathogen that causes febrile illness and can cause chronic arthritis. No approved vaccines or antivirals are available for CHIKV. The attenuation of CHIKV is critical to the establishment of experimental systems that can be used to conduct virus replication studies at a lower biosafety level. We applied a functional selection approach to develop, for the first time, a noncytotoxic CHIKV replicon capable of persisting in human cell lines. We anticipate that this safe and efficient research tool will be valuable for screening CHIKV replication inhibitors and for identifying and analyzing host factors involved in viral replication. We also analyzed, from virological and protein biochemistry perspectives, the functional defects caused by mutations conferring noncytotoxic phenotypes; we found that all known enzymatic activities of CHIKV nsP2, as well as its RNA-binding capability, were compromised by these mutations, which led to a reduced capacity for replication.
Collapse
|
303
|
Chikungunya virus RNA and antibody testing at a National Reference Laboratory since the emergence of Chikungunya virus in the Americas. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 22:291-7. [PMID: 25540275 DOI: 10.1128/cvi.00720-14] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Since first reported in the Americas in December 2013, chikungunya virus (CHIKV) infections have been documented in travelers returning from the Caribbean, with many cases identified by CHIKV antibody and/or RNA testing at our laboratory. We used our large data set to characterize the relationship between antibody titers and RNA detection and to estimate IgM persistence. CHIKV RNA was measured by nucleic acid amplification and CHIKV IgG/IgM by indirect immunofluorescence. Of the 1,306 samples submitted for RNA testing in January through September 2014, 393 (30%) were positive; for 166 RNA-positive samples, CHIKV antibody testing was also ordered, and 84% were antibody negative. Of the 6,971 sera submitted for antibody testing in January through September 2014, 1,811 (26%) were IgM positive; 1,461 IgM positives (81%) were also IgG positive. The relationship between the CHIKV antibody titers and RNA detection was evaluated using 376 IgM-positive samples (138 with RNA testing ordered and 238 deidentified and tested for RNA). RNA detection showed no significant association with the IgM titer but was inversely related to the IgG titer; 63% of the IgG negative sera were RNA positive, compared to 36% of sera with low IgG titers (1:10 to 1:80) and 16% with IgG titers of ≥1:160. Using second-sample results from 62 seroconverters, we estimated that CHIKV IgM persists for 110 days (95% confidence interval, 78 to 150 days) after the initial antibody-negative sample. These findings indicate that (i) RNA detection is more sensitive than antibody detection early in CHIKV infection, (ii) in the absence of RNA results, the IgG titer of the IgM-positive samples may be a useful surrogate for viremia, and (iii) CHIKV IgM persists for approximately 4 months after symptom onset.
Collapse
|
304
|
Abstract
A large epidemic of Chikungunya fever currently affects the Caribbean, Central and South America. Despite a high number of reported cases, little is known on the occurrence of severe clinical complications. We describe four Venezuelan patients with a severe and/or lethal course who exhibit unusual manifestations of the disease. Case 1 describes a 75 year-old man with rapid onset of septic shock and multi-organ failure. Cases 2 and 3 describe two patients with rapid aggressive clinical course who developed shock, severe purpuric lesions and a distinct area large of necrosis in the nasal region. Case 4 depicts a splenectomized woman with shock, generalized purpuric lesions, bullous dermatosis and acronecrosis of an upper limb. Chikungunya fever in the Western hemisphere may also associate with atypical and severe manifestations. Some patients experience a life-threatening, aggressive clinical course, with rapid deterioration and death due to multisystem failure.
Collapse
|
305
|
Burrack KS, Montgomery SA, Homann D, Morrison TE. CD8+ T cells control Ross River virus infection in musculoskeletal tissues of infected mice. THE JOURNAL OF IMMUNOLOGY 2014; 194:678-89. [PMID: 25488988 DOI: 10.4049/jimmunol.1401833] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Ross River virus (RRV), chikungunya virus, and related alphaviruses cause debilitating polyarthralgia and myalgia. Mouse models of RRV and chikungunya virus have demonstrated a role for the adaptive immune response in the control of these infections. However, questions remain regarding the role for T cells in viral control, including the magnitude, location, and dynamics of CD8(+) T cell responses. To address these questions, we generated a recombinant RRV expressing the H-2(b)-restricted glycoprotein 33 (gp33) determinant derived from the glycoprotein of lymphocytic choriomeningitis virus. Using tetramers, we tracked gp33-specific CD8(+) T cells during RRV-lymphocytic choriomeningitis virus infection. We found that acute RRV infection induces activation of CD8(+) T cell responses in lymphoid and musculoskeletal tissues that peak from 10-14 d postinoculation, suggesting that CD8(+) T cells contribute to control of acute RRV infection. Mice genetically deficient for CD8(+) T cells or wild-type mice depleted of CD8(+) T cells had elevated RRV loads in skeletal muscle tissue, but not joint-associated tissues, at 14 d postinoculation, suggesting that the ability of CD8(+) T cells to control RRV infection is tissue dependent. Finally, adoptively transferred T cells were capable of reducing RRV loads in skeletal muscle tissue of Rag1(-/-) mice, indicating that T cells can contribute to the control of RRV infection in the absence of B cells and Ab. Collectively, these data demonstrate a role for T cells in the control of RRV infection and suggest that the antiviral capacity of T cells is controlled in a tissue-specific manner.
Collapse
Affiliation(s)
- Kristina S Burrack
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045
| | - Stephanie A Montgomery
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Dirk Homann
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045; Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO 80045; Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Thomas E Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045;
| |
Collapse
|
306
|
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
|
307
|
Lo Presti A, Lai A, Cella E, Zehender G, Ciccozzi M. Chikungunya virus, epidemiology, clinics and phylogenesis: A review. ASIAN PAC J TROP MED 2014; 7:925-32. [PMID: 25479619 DOI: 10.1016/s1995-7645(14)60164-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 07/15/2014] [Accepted: 10/15/2014] [Indexed: 11/28/2022] Open
Abstract
Chikungunya virus is a mosquito-transmitted alphavirus that causes chikungunya fever, a febrile illness associated with severe arthralgia and rash. Chikungunya virus is transmitted by culicine mosquitoes; Chikungunya virus replicates in the skin, disseminates to liver, muscle, joints, lymphoid tissue and brain, presumably through the blood. Phylogenetic studies showed that the Indian Ocean and the Indian subcontinent epidemics were caused by two different introductions of distinct strains of East/Central/South African genotype of CHIKV. The paraphyletic grouping of African CHIK viruses supports the historical evidence that the virus was introduced into Asia from Africa. Phylogenetic analysis divided Chikungunya virus isolates into three distinct genotypes based on geographical origins: the first, the West Africa genotype, consisted of isolates from Senegal and Nigeria; the second contained strains from East/Central/South African genotype, while the third contained solely Asian. The most recent common ancestor for the recent epidemic, which ravaged Indian Ocean islands and Indian subcontinent in 2004 - 2007, was found to date in 2002. Asian lineage dated about 1952 and exhibits similar spread patterns of the recent Indian Ocean outbreak lineage, with successive epidemics detected along an eastward path. Asian group splitted into two clades: an Indian lineage and a south east lineage. Outbreaks of Chikungunya virus fever in Asia have not been associated necessarily with outbreaks in Africa. Phylogenetic tools can reconstruct geographic spread of Chikungunya virus during the epidemics wave. The good management of patients with acute Chikungunya virus infection is essential for public health in susceptible areas with current Aedes spp activity.
Collapse
Affiliation(s)
- Alessandra Lo Presti
- Department of Infectious Parasitic and Immunomediated Diseases, Epidemiology Unit, Reference Centre on Phylogeny, Molecular Epidemiology and Microbial Evolution (FEMEM), Istituto Superiore di Sanita', Rome, Italy
| | - Alessia Lai
- Department of Biomedical and Clinical Sciences, L. Sacco Hospital, University of Milan, Milan, Italy
| | - Eleonora Cella
- Department of Infectious Parasitic and Immunomediated Diseases, Epidemiology Unit, Reference Centre on Phylogeny, Molecular Epidemiology and Microbial Evolution (FEMEM), Istituto Superiore di Sanita', Rome, Italy
| | - Gianguglielmo Zehender
- Department of Biomedical and Clinical Sciences, L. Sacco Hospital, University of Milan, Milan, Italy
| | - Massimo Ciccozzi
- Department of Infectious Parasitic and Immunomediated Diseases, Epidemiology Unit, Reference Centre on Phylogeny, Molecular Epidemiology and Microbial Evolution (FEMEM), Istituto Superiore di Sanita', Rome, Italy; University Campus-Biomedico, Rome, Italy.
| |
Collapse
|
308
|
Nuckols JT, McAuley AJ, Huang YJS, Horne KM, Higgs S, Davey RA, Vanlandingham DL. pH-Dependent entry of chikungunya virus fusion into mosquito cells. Virol J 2014; 11:215. [PMID: 25476236 PMCID: PMC4266220 DOI: 10.1186/s12985-014-0215-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Accepted: 11/25/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Millions of human infections caused by arthropod-borne pathogens are initiated by the feeding of an infected mosquito on a vertebrate. However, interactions between the viruses and the mosquito vector, which facilitates successful infection and transmission of virus to a subsequent vertebrate host, are still not fully understood. FINDING Here we describe early chikungunya virus (CHIKV) infectious events in cells derived from one of the most important CHIKV vectors, Aedes albopictus. We demonstrated that CHIKV infection of mosquito cells depended on acidification of the endosome as indicated by significant inhibition following prophylactic treatment with the lysosomotropic drugs chloroquine, ammonium chloride, and monensin, which is consistent with observations in mammalian cells. While all three agents inhibited CHIKV infection in C6/36 cells, ammonium chloride was less toxic to cells than the other agents. CONCLUSION The observation of similar mechanisms for inhibition of CHIKV infection in mosquito and mammalian cell lines suggests that conserved entry pathways are utilized by CHIKV for vertebrate and invertebrate cell types.
Collapse
Affiliation(s)
- John T Nuckols
- Medical Countermeasure Systems, Joint Vaccine Acquisition Program, Fort Detrick, MD, 21702, USA.
| | - Alexander J McAuley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, 77555, USA.
| | - Yan-Jang S Huang
- Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, 66506, USA. .,Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, 66506, USA.
| | - Kate M Horne
- Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, 66506, USA.
| | - Stephen Higgs
- Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, 66506, USA. .,Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, 66506, USA.
| | - Robert A Davey
- Texas Biomedical Research Institute, San Antonio, TX, 78245, USA.
| | - Dana L Vanlandingham
- Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, 66506, USA. .,Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|
309
|
Poo YS, Rudd PA, Gardner J, Wilson JAC, Larcher T, Colle MA, Le TT, Nakaya HI, Warrilow D, Allcock R, Bielefeldt-Ohmann H, Schroder WA, Khromykh AA, Lopez JA, Suhrbier A. Multiple immune factors are involved in controlling acute and chronic chikungunya virus infection. PLoS Negl Trop Dis 2014; 8:e3354. [PMID: 25474568 PMCID: PMC4256279 DOI: 10.1371/journal.pntd.0003354] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 10/15/2014] [Indexed: 12/18/2022] Open
Abstract
The recent epidemic of the arthritogenic alphavirus, chikungunya virus (CHIKV) has prompted a quest to understand the correlates of protection against virus and disease in order to inform development of new interventions. Herein we highlight the propensity of CHIKV infections to persist long term, both as persistent, steady-state, viraemias in multiple B cell deficient mouse strains, and as persistent RNA (including negative-strand RNA) in wild-type mice. The knockout mouse studies provided evidence for a role for T cells (but not NK cells) in viraemia suppression, and confirmed the role of T cells in arthritis promotion, with vaccine-induced T cells also shown to be arthritogenic in the absence of antibody responses. However, MHC class II-restricted T cells were not required for production of anti-viral IgG2c responses post CHIKV infection. The anti-viral cytokines, TNF and IFNγ, were persistently elevated in persistently infected B and T cell deficient mice, with adoptive transfer of anti-CHIKV antibodies unable to clear permanently the viraemia from these, or B cell deficient, mice. The NOD background increased viraemia and promoted arthritis, with B, T and NK deficient NOD mice showing high-levels of persistent viraemia and ultimately succumbing to encephalitic disease. In wild-type mice persistent CHIKV RNA and negative strand RNA (detected for up to 100 days post infection) was associated with persistence of cellular infiltrates, CHIKV antigen and stimulation of IFNα/β and T cell responses. These studies highlight that, secondary to antibodies, several factors are involved in virus control, and suggest that chronic arthritic disease is a consequence of persistent, replicating and transcriptionally active CHIKV RNA. The largest epidemic ever recorded for chikungunya virus (CHIKV) started in 2004 in Africa, then spread across Asia and recently caused tens of thousands of cases in Papua New Guinea and the Caribbean. This mosquito-borne alphavirus primarily causes an often debilitating, acute and chronic polyarthritis/polyarthalgia. Despite robust anti-viral immune responses CHIKV is able to persist, with such persistence poorly understood and the likely cause of chronic disease. Herein we highlight the propensity of CHIKV to persist long term, both as a persistent viraemia in different B cell deficient mouse strains, but also as persistent viral RNA in wild-type mice. These studies suggest that, aside from antibodies, other immune factors, such as CD4 T cells and TNF, are active in viraemia control. The work also supports the notion that CHIKV disease, with the exception of encephalitis, is largely an immunopathology. Persistent CHIKV RNA in wild-type mice continues to stimulate type I interferon and T cell responses, with this model of chronic disease recapitulating many of the features seen in chronic CHIKV patients.
Collapse
Affiliation(s)
- Yee Suan Poo
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
- School of Medicine/School of Molecular and Microbial Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Penny A. Rudd
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
- School of Medicine/School of Molecular and Microbial Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Joy Gardner
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
| | - Jane A. C. Wilson
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
- School of Medicine/School of Molecular and Microbial Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Thibaut Larcher
- Institut National de Recherche Agronomique, Unité Mixte de Recherche 703, Oniris, Nantes, France
| | - Marie-Anne Colle
- Institut National de Recherche Agronomique, Unité Mixte de Recherche 703, Oniris, Nantes, France
| | - Thuy T. Le
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
| | - Helder I. Nakaya
- School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - David Warrilow
- Public Health Virology Laboratory, Department of Health, Queensland Government, Brisbane, Queensland, Australia
| | - Richard Allcock
- Lotterywest State Biomedical Facility Genomics, Royal Perth Hospital, Perth, Western Australia, Australia
| | | | - Wayne A. Schroder
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
| | - Alexander A. Khromykh
- School of Medicine/School of Molecular and Microbial Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - José A. Lopez
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
- School of Natural Sciences, Griffith University, Nathan, Australia
| | - Andreas Suhrbier
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
- School of Medicine/School of Molecular and Microbial Sciences, University of Queensland, Brisbane, Queensland, Australia
- School of Natural Sciences, Griffith University, Nathan, Australia
- * E-mail:
| |
Collapse
|
310
|
Coffey LL, Failloux AB, Weaver SC. Chikungunya virus-vector interactions. Viruses 2014; 6:4628-63. [PMID: 25421891 PMCID: PMC4246241 DOI: 10.3390/v6114628] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/10/2014] [Accepted: 11/10/2014] [Indexed: 12/25/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that causes chikungunya fever, a severe, debilitating disease that often produces chronic arthralgia. Since 2004, CHIKV has emerged in Africa, Indian Ocean islands, Asia, Europe, and the Americas, causing millions of human infections. Central to understanding CHIKV emergence is knowledge of the natural ecology of transmission and vector infection dynamics. This review presents current understanding of CHIKV infection dynamics in mosquito vectors and its relationship to human disease emergence. The following topics are reviewed: CHIKV infection and vector life history traits including transmission cycles, genetic origins, distribution, emergence and spread, dispersal, vector competence, vector immunity and microbial interactions, and co-infection by CHIKV and other arboviruses. The genetics of vector susceptibility and host range changes, population heterogeneity and selection for the fittest viral genomes, dual host cycling and its impact on CHIKV adaptation, viral bottlenecks and intrahost diversity, and adaptive constraints on CHIKV evolution are also discussed. The potential for CHIKV re-emergence and expansion into new areas and prospects for prevention via vector control are also briefly reviewed.
Collapse
Affiliation(s)
- Lark L Coffey
- Center for Vectorborne Diseases, School of Veterinary Medicine, University of California, Davis, CA 95616, USA.
| | - Anna-Bella Failloux
- Department of Virology, Arboviruses and Insect Vectors, Institut Pasteur, 25-28 rue du Dr. Roux, 75724 Paris cedex 15, France.
| | - Scott C Weaver
- Institute for Human Infections and Immunity, Center for Tropical Diseases and Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
311
|
Bhakat S, Karubiu W, Jayaprakash V, Soliman ME. A perspective on targeting non-structural proteins to combat neglected tropical diseases: Dengue, West Nile and Chikungunya viruses. Eur J Med Chem 2014; 87:677-702. [DOI: 10.1016/j.ejmech.2014.10.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 09/29/2014] [Accepted: 10/04/2014] [Indexed: 01/07/2023]
|
312
|
Taylor A, Herrero LJ, Rudd PA, Mahalingam S. Mouse models of alphavirus-induced inflammatory disease. J Gen Virol 2014; 96:221-238. [PMID: 25351726 DOI: 10.1099/vir.0.071282-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Part of the Togaviridae family, alphaviruses are arthropod-borne viruses that are widely distributed throughout the globe. Alphaviruses are able to infect a variety of vertebrate hosts, but in humans, infection can result in extensive morbidity and mortality. Symptomatic infection can manifest as fever, an erythematous rash and/or significant inflammatory pathologies such as arthritis and encephalitis. Recent overwhelming outbreaks of alphaviral disease have highlighted the void in our understanding of alphavirus pathogenesis and the re-emergence of alphaviruses has given new impetus to anti-alphaviral drug design. In this review, the development of viable mouse models of Old Word and New World alphaviruses is examined. How mouse models that best replicate human disease have been used to elucidate the immunopathology of alphavirus pathogenesis and trial novel therapeutic discoveries is also discussed.
Collapse
Affiliation(s)
- Adam Taylor
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, Australia
| | - Lara J Herrero
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, Australia
| | - Penny A Rudd
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, Australia
| | - Suresh Mahalingam
- Institute for Glycomics, Griffith University, Gold Coast Campus, QLD, Australia
| |
Collapse
|
313
|
Das T, Hoarau JJ, Bandjee MCJ, Maquart M, Gasque P. Multifaceted innate immune responses engaged by astrocytes, microglia and resident dendritic cells against Chikungunya neuroinfection. J Gen Virol 2014; 96:294-310. [PMID: 25351727 DOI: 10.1099/vir.0.071175-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Chikungunya virus (CHIKV) has recently affected millions of people in the Indian Ocean, with rare cases of encephalopathy and encephalitis occurring in neonates. In the study described herein, the capacity of mouse brain cells to control infection through innate immune antiviral responses was assessed. In vitro, CHIKV principally infected a subpopulation of mouse GFAP+ primary astrocytes. Oligodendrocytes and neurons could also be infected. An innate immune response was engaged by CHIKV-infected astrocytes with elevated expression of mRNAs for IFN-α-β, inflammatory cytokines (e.g. IL-1β, IL-12, IL-10, IL-24) and proapoptotic factors (e.g. TNF-α, FasL, Lymphotoxin B). Programmed cell death through the intrinsic caspase-9 pathway was observed by immunofluorescence in infected astrocytes and neurons but not in oligodendrocytes. Interestingly, microglia did not replicate CHIKV but responded by elevated mitogen-activated protein kinase (MAPK) activity. Intracerebroventricular injection of CHIKV in neonate mice led to the infection of astrocytes. The astrogliosis response was accompanied by a dendritic CD206+ cell mobilization restricted to the site of infection. The results of this study support the paradigm that a multifaceted innate immune response can be mobilized by both professional immune and glial cells to control CHIKV neuroinfection events in neonates.
Collapse
Affiliation(s)
- Trina Das
- Immunopathology and Infection Research Grouping (IRG, GRI EA4517), University of la Reunion, CHU and CYROI, St Denis, Reunion Island
| | - Jean Jacques Hoarau
- Immunopathology and Infection Research Grouping (IRG, GRI EA4517), University of la Reunion, CHU and CYROI, St Denis, Reunion Island
| | - Marie Christine Jaffar Bandjee
- Virology laboratory, CHU Félix Guyon of la Réunion, St Denis, Reunion Island.,Immunopathology and Infection Research Grouping (IRG, GRI EA4517), University of la Reunion, CHU and CYROI, St Denis, Reunion Island
| | - Marianne Maquart
- CIRAD and CYROI, St Denis, Reunion Island.,CRVOI, St Denis, Reunion Island
| | - Philippe Gasque
- Immunopathology and Infection Research Grouping (IRG, GRI EA4517), University of la Reunion, CHU and CYROI, St Denis, Reunion Island
| |
Collapse
|
314
|
Rougeron V, Sam IC, Caron M, Nkoghe D, Leroy E, Roques P. Chikungunya, a paradigm of neglected tropical disease that emerged to be a new health global risk. J Clin Virol 2014; 64:144-52. [PMID: 25453326 DOI: 10.1016/j.jcv.2014.08.032] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 08/25/2014] [Indexed: 01/08/2023]
Abstract
Chikungunya virus (CHIKV) is an alphavirus of the Togaviridae family that causes chronic and incapacitating arthralgia in human populations. Since its discovery in 1952, CHIKV was responsible for sporadic and infrequent outbreaks. However, since 2005, global Chikungunya outbreaks have occurred, inducing some fatalities and associated with severe and chronic morbidity. Chikungunya is thus considered as an important re-emerging public health problem in both tropical and temperate countries, where the distribution of the Aedes mosquito vectors continues to expand. This review highlights the most recent advances in our knowledge and understanding of the epidemiology, biology, treatment and vaccination strategies of CHIKV.
Collapse
Affiliation(s)
- Virginie Rougeron
- Centre International de Recherches Médicales de Franceville, Franceville, Gabon; Unité Mixte de Recherche Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle (IRD 224 - CNRS 5290 - UM1-UM2), Institut de Recherche pour le Développement, Montpellier, France
| | - I-Ching Sam
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mélanie Caron
- Centre International de Recherches Médicales de Franceville, Franceville, Gabon
| | - Dieudonné Nkoghe
- Centre International de Recherches Médicales de Franceville, Franceville, Gabon
| | - Eric Leroy
- Centre International de Recherches Médicales de Franceville, Franceville, Gabon; Unité Mixte de Recherche Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle (IRD 224 - CNRS 5290 - UM1-UM2), Institut de Recherche pour le Développement, Montpellier, France
| | - Pierre Roques
- CEA, Institute of Emerging Diseases and Innovative Therapies, Division of Immuno-Virology, Fontenay-aux-Roses, France; Université Paris-Sud 11, UMR E1, Orsay, France.
| |
Collapse
|
315
|
Nitatpattana N, Kanjanopas K, Yoksan S, Satimai W, Vongba N, Langdatsuwan S, Nakgoi K, Ratchakum S, Wauquier N, Souris M, Auewarakul P, Gonzalez JP. Long-term persistence of Chikungunya virus neutralizing antibodies in human populations of North Eastern Thailand. Virol J 2014; 11:183. [PMID: 25330992 PMCID: PMC4283153 DOI: 10.1186/1743-422x-11-183] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 09/18/2014] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Chikungunya virus (CHIKV) outbreak recurrences in Thailand are unpredictable and separated by unexplained and often long silent epidemiological periods that can last for several years. These silent periods could be explained in part by the fact that infection with one CHIKV strain confers lasting natural immunity, even against other CHIKV strains. In this study we evaluated the persistence of CHIKV-specific neutralizing antibodies in the population of Chumpae District, Khon Kaen Province, nineteen years after a CHIKV outbreak occurred in the same area in 1991. FINDINGS Overall 39% (44/111) of 111 former patients had neutralizing antibodies reacting against CHIKV ECSA strain. Consistently high titers of neutralizing antibodies were found in 75% (33/44) of all positively-reacting sera, 70% of which (23/33) were collected from individuals amongst the >60 years old age group. Although the prevalence found in Pong Haeng village (70%) was significantly higher than the prevalence detected in the Nong Thum village (14%), control study villages without known previous Chikungunya epidemics had a high Chikungunya neutralizing antibody prevalence (65%). CONCLUSIONS More than one-third of the pre-exposed population had persisting natural immunity that was more likely boosted by recent and repetitive exposure to the emerging ECSA CHIKV in Thailand. Also, Chikungunya virus appears to largely circulate in the country with a great variability appears between villages or area probably associated with the vector abundance and efficiency. Altogether these results show a potential for a lifelong immunity against CHIKV. Given the rapid spread of the highly pathogenic ECSA strain in Southern Thailand, the development of CHIK vaccine is strongly recommended.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jean-Paul Gonzalez
- Institute of Molecular Bioscience, Mahidol University, Nakhon Pathom, Thailand.
| |
Collapse
|
316
|
Petersen LR, Epstein JS. Chikungunya virus: new risk to transfusion safety in the Americas. Transfusion 2014; 54:1911-5. [PMID: 25130331 DOI: 10.1111/trf.12790] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Lyle R Petersen
- Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Fort Collins, CO.
| | | |
Collapse
|
317
|
Christofferson RC, Chisenhall DM, Wearing HJ, Mores CN. Chikungunya viral fitness measures within the vector and subsequent transmission potential. PLoS One 2014; 9:e110538. [PMID: 25310016 PMCID: PMC4195746 DOI: 10.1371/journal.pone.0110538] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/06/2014] [Indexed: 12/23/2022] Open
Abstract
Given the recent emergence of chikungunya in the Americas, the accuracy of forecasting and prediction of chikungunya transmission potential in the U.S. requires urgent assessment. The La Reunion-associated sub-lineage of chikungunya (with a valine substitution in the envelope protein) was shown to increase viral fitness in the secondary vector, Ae. albopictus. Subsequently, a majority of experimental and modeling efforts focused on this combination of a sub-lineage of the East-Central-South African genotype (ECSA-V) – Ae. albopictus, despite the Asian genotype being the etiologic agent of recent chikungunya outbreaks world-wide. We explore a collection of data to investigate relative transmission efficiencies of the three major genotypes/sub-lineages of chikungunya and found difference in the extrinsic incubation periods to be largely overstated. However, there is strong evidence supporting the role of Ae. albopictus in the expansion of chikungunya that our R0 calculations cannot attribute to fitness increases in one vector over another. This suggests other ecological factors associated with the Ae. albopictus-ECSA-V cycle may drive transmission intensity differences. With the apparent bias in literature, however, we are less prepared to evaluate transmission where Ae. aegypti plays a significant role. Holistic investigations of CHIKV transmission cycle(s) will allow for more complete assessment of transmission risk in areas affected by either or both competent vectors.
Collapse
Affiliation(s)
- Rebecca C. Christofferson
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, United States of America
- * E-mail:
| | - Daniel M. Chisenhall
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Helen J. Wearing
- Department of Biology, The University of New Mexico, Albuquerque, New Mexico, United States of America
- Department of Mathematics & Statistics, The University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Christopher N. Mores
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, United States of America
| |
Collapse
|
318
|
Exposure of epitope residues on the outer face of the chikungunya virus envelope trimer determines antibody neutralizing efficacy. J Virol 2014; 88:14364-79. [PMID: 25275138 DOI: 10.1128/jvi.01943-14] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED Chikungunya virus (CHIKV) is a reemerging alphavirus that causes a debilitating arthritic disease and infects millions of people and for which no specific treatment is available. Like many alphaviruses, the structural targets on CHIKV that elicit a protective humoral immune response in humans are poorly defined. Here we used phage display against virus-like particles (VLPs) to isolate seven human monoclonal antibodies (MAbs) against the CHIKV envelope glycoproteins E2 and E1. One MAb, IM-CKV063, was highly neutralizing (50% inhibitory concentration, 7.4 ng/ml), demonstrated high-affinity binding (320 pM), and was capable of therapeutic and prophylactic protection in multiple animal models up to 24 h postexposure. Epitope mapping using a comprehensive shotgun mutagenesis library of 910 mutants with E2/E1 alanine mutations demonstrated that IM-CKV063 binds to an intersubunit conformational epitope on domain A, a functionally important region of E2. MAbs against the highly conserved fusion loop have not previously been reported but were also isolated in our studies. Fusion loop MAbs were broadly cross-reactive against diverse alphaviruses but were nonneutralizing. Fusion loop MAb reactivity was affected by temperature and reactivity conditions, suggesting that the fusion loop is hidden in infectious virions. Visualization of the binding sites of 15 different MAbs on the structure of E2/E1 revealed that all epitopes are located at the membrane-distal region of the E2/E1 spike. Interestingly, epitopes on the exposed topmost and outer surfaces of the E2/E1 trimer structure were neutralizing, whereas epitopes facing the interior of the trimer were not, providing a rationale for vaccine design and therapeutic MAb development using the intact CHIKV E2/E1 trimer. IMPORTANCE CHIKV is the most important alphavirus affecting humans, resulting in a chronic arthritic condition that can persist for months or years. In recent years, millions of people have been infected globally, and the spread of CHIKV to the Americas is now beginning, with over 100,000 cases occurring in the Caribbean within 6 months of its arrival. Our study reports on seven human MAbs against the CHIKV envelope, including a highly protective MAb and rarely isolated fusion loop MAbs. Epitope mapping of these MAbs demonstrates how some E2/E1 epitopes are exposed or hidden from the human immune system and suggests a structural mechanism by which these MAbs protect (or fail to protect) against CHIKV infection. Our results suggest that the membrane-distal end of CHIKV E2/E1 is the primary target for the humoral immune response to CHIKV, and antibodies targeting the exposed topmost and outer surfaces of the E2/E1 trimer determine the neutralizing efficacy of this response.
Collapse
|
319
|
Owino EA, Sang R, Sole CL, Pirk C, Mbogo C, Torto B. Field evaluation of natural human odours and the biogent-synthetic lure in trapping Aedes aegypti, vector of dengue and chikungunya viruses in Kenya. Parasit Vectors 2014; 7:451. [PMID: 25246030 PMCID: PMC4261536 DOI: 10.1186/1756-3305-7-451] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 09/15/2014] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Methods currently used in sampling adult Aedes aegypti, the main vector of dengue and chikungunya viruses are limited for effective surveillance of the vector and accurate determination of the extent of virus transmission during outbreaks and inter - epidemic periods. Here, we document the use of natural human skin odours in baited traps to improve sampling of adult Ae. aegypti in two different endemic areas of chikungunya and dengue in Kenya - Kilifi and Busia Counties. The chemistry of the volatiles released from human odours and the Biogent (BG)-commercial lure were also compared. METHODS Cotton socks and T-shirts were used to obtain natural human skin volatiles from the feet and trunk of three volunteers (volunteers 1 and 2 in Kilifi and volunteers 2 and 3 in Busia). Using Latin square design, we compared the efficacies of BG sentinel traps baited with carbon dioxide plus (a) no bait, (b) human feet volatiles, (c) human trunk volatiles each against (c) a control (Biogent commercial lure) at the two sites. Coupled gas chromatography-mass spectrometry (GC-MS) was used to identify and compare candidate attractants released by the commercial lure and human odours. RESULTS Ae. aegypti captured in the trap baited with feet odours from volunteer 2 and trunk odours from the same volunteer were significantly higher than in the control trap in Busia and Kilifi respectively, [IRR = 5.63, 95% CI: 1.15 - 28.30, p = 0.030] and [IRR = 3.99, 95% CI: 0.95-16.69, p = 0.049]. At both sites, Ae. aegypti captures in traps baited with either the feet or trunk odours from volunteers 1 and 3 were not significantly different from the control. Major qualitative differences were observed between the chemical profiles of human odours and the commercial BG-lure. Aldehydes, fatty acids and ketones dominated human odour profiles, whereas the BG-lure released mainly hexanoic acid. CONCLUSIONS Our results suggest that additional candidate attractants are present in human skin volatiles which can help to improve the efficacy of lures for trapping and surveillance of Ae. aegypti.
Collapse
Affiliation(s)
- Eunice A Owino
- />International Centre of Insect Physiology and Ecology, P.O BOX 30772–00100, Nairobi, Kenya
- />Department of Zoology and Entomology, University of Pretoria, Pretoria, South Africa
| | - Rosemary Sang
- />International Centre of Insect Physiology and Ecology, P.O BOX 30772–00100, Nairobi, Kenya
- />Centre for Virus Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Catherine L Sole
- />Department of Zoology and Entomology, University of Pretoria, Pretoria, South Africa
| | - Christian Pirk
- />Department of Zoology and Entomology, University of Pretoria, Pretoria, South Africa
| | - Charles Mbogo
- />Centre for Geographic Medicine Research – Coast, KEMRI & KEMRI – Wellcome Trust Research Programme, Kilifi, Kenya
| | - Baldwyn Torto
- />International Centre of Insect Physiology and Ecology, P.O BOX 30772–00100, Nairobi, Kenya
| |
Collapse
|
320
|
Robinson M, Conan A, Duong V, Ly S, Ngan C, Buchy P, Tarantola A, Rodó X. A model for a chikungunya outbreak in a rural Cambodian setting: implications for disease control in uninfected areas. PLoS Negl Trop Dis 2014; 8:e3120. [PMID: 25210729 PMCID: PMC4161325 DOI: 10.1371/journal.pntd.0003120] [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: 09/19/2013] [Accepted: 07/15/2014] [Indexed: 11/21/2022] Open
Abstract
Following almost 30 years of relative silence, chikungunya fever reemerged in Kenya in 2004. It subsequently spread to the islands of the Indian Ocean, reaching Southeast Asia in 2006. The virus was first detected in Cambodia in 2011 and a large outbreak occurred in the village of Trapeang Roka Kampong Speu Province in March 2012, in which 44% of the villagers had a recent infection biologically confirmed. The epidemic curve was constructed from the number of biologically-confirmed CHIKV cases per day determined from the date of fever onset, which was self-reported during a data collection campaign conducted in the village after the outbreak. All individuals participating in the campaign had infections confirmed by laboratory analysis, allowing for the identification of asymptomatic cases and those with an unreported date of fever onset. We develop a stochastic model explicitly including such cases, all of whom do not appear on the epidemic curve. We estimate the basic reproduction number of the outbreak to be 6.46 (95% C.I. [6.24, 6.78]). We show that this estimate is particularly sensitive to changes in the biting rate and mosquito longevity. Our model also indicates that the infection was more widespread within the population on the reported epidemic start date. We show that the exclusion of asymptomatic cases and cases with undocumented onset dates can lead to an underestimation of the reproduction number which, in turn, could negatively impact control strategies implemented by public health authorities. We highlight the need for properly documenting newly emerging pathogens in immunologically naive populations and the importance of identifying the route of disease introduction. During the recent resurgence of chikungunya, the scale of imported cases into previously unaffected countries has caused great concern due to the presence of a competent vector (Aedes albopictus) in many of these regions. This study describes a mathematical model for a chikungunya outbreak in the rural Cambodian village of Trapeang Roka, where a chikungunya epidemic was recorded and documented in March 2012. The outbreak data is unique, in that all infections were confirmed by laboratory analysis, enabling the identification of asymptomatic individuals, in addition to individuals who failed to report details of their infection. A stochastic model, partitioning the infectious population into three distinct classes, is implemented using Gillespie's algorithm. We show that the incorporation of both biologically-confirmed symptomatic cases undocumented by date of fever onset and asymptomatic cases yields a higher estimate of the reproduction number. Our results highlight how reproduction numbers could be underestimated by limiting analysis to the epidemic curve. Carefully documenting cases and performing laboratory testing in cluster regions, such as the village considered here, could provide a more comprehensive insight into the true infection dynamics.
Collapse
Affiliation(s)
| | - Anne Conan
- Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Veasna Duong
- Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Sowath Ly
- Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Chantha Ngan
- National Dengue Control Program, Ministry of Health, Phnom Penh, Cambodia
| | | | | | - Xavier Rodó
- Institut Català de Ciències del Clima, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
321
|
Involvement of ATP synthase β subunit in chikungunya virus entry into insect cells. Arch Virol 2014; 159:3353-64. [PMID: 25168043 DOI: 10.1007/s00705-014-2210-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 08/20/2014] [Indexed: 10/24/2022]
Abstract
Chikungunya virus (CHIKV), the virus responsible for the disease chikungunya fever in humans, is transmitted by Aedes mosquitoes. While significant progress has been made in understanding the process by which CHIKV enters into mammalian cells, far less progress has been made in understanding the CHIKV entry process in insect cells. This study sought to identify mosquito-cell-expressed CHIKV-binding proteins through a combination of virus overlay protein binding assays (VOPBA) and mass spectroscopy. A 50-kDa CHIKV-binding protein was identified as the ATP synthase β subunit (ATPSβ). Co-immunoprecipitation studies confirmed the interaction, and colocalization analysis showed cell-surface and intracellular co-localization between CHIKV and ATPSβ. Both antibody inhibition and siRNA-mediated downregulation experiments targeted to ATPSβ showed a significant reduction in viral entry and virus production. These results suggest that ATPSβ is a CHIKV-binding protein capable of mediating the entry of CHIKV into insect cells.
Collapse
|
322
|
Jupille H, Vega-Rua A, Rougeon F, Failloux AB. Arboviruses: variations on an ancient theme. Future Virol 2014. [DOI: 10.2217/fvl.14.62] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
ABSTRACT Arboviruses utilize different strategies to complete their transmission cycle between vertebrate and invertebrate hosts. Most possess an RNA genome coupled with an RNA polymerase lacking proofreading activity and generate large populations of genetically distinct variants, permitting rapid adaptation to environmental changes. With mutation rates of between 10- 6 and 10-4 substitutions per nucleotide, arboviral genomes rapidly acquire mutations that can lead to viral emergence. Arboviruses can be described in seven families, four of which have medical importance: Togaviridae, Flaviviridae, Bunyaviridae and Reoviridae. The Togaviridae and Flaviviridae both have ssRNA genomes, while the Bunyaviridae and Reoviridae possess segmented RNA genomes. Recent epidemics caused by these arboviruses have been associated with specific mutations leading to enhanced host ranges, vector shifts and virulence.
Collapse
Affiliation(s)
- Henri Jupille
- Department of Virology, Arboviruses & Insect Vectors, 25 Rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Anubis Vega-Rua
- Department of Virology, Arboviruses & Insect Vectors, 25 Rue du Dr Roux, 75724 Paris Cedex 15, France
- Cellule Pasteur UPMC, Université Pierre et Marie Curie, Paris, France
| | | | - Anna-Bella Failloux
- Department of Virology, Arboviruses & Insect Vectors, 25 Rue du Dr Roux, 75724 Paris Cedex 15, France
| |
Collapse
|
323
|
Tan JJL, Capozzoli M, Sato M, Watthanaworawit W, Ling CL, Mauduit M, Malleret B, Grüner AC, Tan R, Nosten FH, Snounou G, Rénia L, Ng LFP. An integrated lab-on-chip for rapid identification and simultaneous differentiation of tropical pathogens. PLoS Negl Trop Dis 2014; 8:e3043. [PMID: 25078474 PMCID: PMC4117454 DOI: 10.1371/journal.pntd.0003043] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 06/10/2014] [Indexed: 01/03/2023] Open
Abstract
Tropical pathogens often cause febrile illnesses in humans and are responsible for considerable morbidity and mortality. The similarities in clinical symptoms provoked by these pathogens make diagnosis difficult. Thus, early, rapid and accurate diagnosis will be crucial in patient management and in the control of these diseases. In this study, a microfluidic lab-on-chip integrating multiplex molecular amplification and DNA microarray hybridization was developed for simultaneous detection and species differentiation of 26 globally important tropical pathogens. The analytical performance of the lab-on-chip for each pathogen ranged from 102 to 103 DNA or RNA copies. Assay performance was further verified with human whole blood spiked with Plasmodium falciparum and Chikungunya virus that yielded a range of detection from 200 to 4×105 parasites, and from 250 to 4×107 PFU respectively. This lab-on-chip was subsequently assessed and evaluated using 170 retrospective patient specimens in Singapore and Thailand. The lab-on-chip had a detection sensitivity of 83.1% and a specificity of 100% for P. falciparum; a sensitivity of 91.3% and a specificity of 99.3% for P. vivax; a positive 90.0% agreement and a specificity of 100% for Chikungunya virus; and a positive 85.0% agreement and a specificity of 100% for Dengue virus serotype 3 with reference methods conducted on the samples. Results suggested the practicality of an amplification microarray-based approach in a field setting for high-throughput detection and identification of tropical pathogens. Tropical diseases consist of a group of debilitating and fatal infections that occur primarily in rural and urban settings of tropical and subtropical countries. While the primary indices of an infection are mostly the presentation of clinical signs and symptoms, outcomes due to an infection with tropical pathogens are often unspecific. Accurate diagnosis is crucial for timely intervention, appropriate and adequate treatments, and patient management to prevent development of sequelae and transmission. Although, multiplex assays are available for the simultaneous detection of tropical pathogens, they are generally of low throughput. Performing parallel assays to cover the detection for a comprehensive scope of tropical infections that include protozoan, bacterial and viral infections is undoubtedly labor-intensive and time consuming. We present an integrated lab-on-chip using microfluidics technology coupled with reverse transcription (RT), PCR amplification, and microarray hybridization for the simultaneous identification and differentiation of 26 tropical pathogens that cause 14 globally important tropical diseases. Such diagnostics capacity would facilitate evidence-based management of patients, improve the specificity of treatment and, in some cases, even allow contact tracing and other disease-control measures.
Collapse
Affiliation(s)
- Jeslin J. L. Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Monica Capozzoli
- CI Group, Molecular Diagnostic Business Unit, Microfluidics Division, ST Microelectronics, Catania, Italy
| | - Mitsuharu Sato
- Veredus Laboratories Pte Ltd, Singapore Science Park, Singapore
| | - Wanitda Watthanaworawit
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Clare L. Ling
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Marjorie Mauduit
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Benoît Malleret
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Anne-Charlotte Grüner
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
| | - Rosemary Tan
- Veredus Laboratories Pte Ltd, Singapore Science Park, Singapore
| | - François H. Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Georges Snounou
- Université Pierre et Marie Curie (Paris VI), Centre Hospitalo-Universitaire Pitié-Salpêtrière, Paris, France
- INSERM UMR S 945, Paris, France
| | - Laurent Rénia
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
- * E-mail: (LR); (LFPN)
| | - Lisa F. P. Ng
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- * E-mail: (LR); (LFPN)
| |
Collapse
|
324
|
Weller N, Clowes P, Dobler G, Saathoff E, Kroidl I, Ntinginya NE, Maboko L, Löscher T, Hoelscher M, Heinrich N. Seroprevalence of alphavirus antibodies in a cross-sectional study in southwestern Tanzania suggests endemic circulation of chikungunya. PLoS Negl Trop Dis 2014; 8:e2979. [PMID: 25079964 PMCID: PMC4117434 DOI: 10.1371/journal.pntd.0002979] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 05/13/2014] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND To date, Alphavirus infections and their most prominent member, chikungunya fever, a viral disease which first became apparent in Tanzania in 1953, have been very little investigated in regions without epidemic occurrence. Few data exist on burden of disease and socio-economic and environmental covariates disposing to infection. METHODS A cross-sectional seroprevalence study was undertaken in 1,215 persons from Mbeya region, South-Western Tanzania, to determine the seroprevalence of anti-Alphavirus IgG antibodies, and to investigate associated risk factors. RESULTS 18% of 1,215 samples were positive for Alphavirus IgG. Seropositivity was associated with participant age, low to intermediate elevation, flat terrain and with IgG positivity for Rift Valley fever, Flaviviridae, and rickettsiae of the spotted fever group. When comparing the geographical distribution of Alphavirus seropositivity to that of Rift Valley fever, it was obvious that Alphaviruses had spread more widely throughout the study area, while Rift Valley fever was concentrated along the shore of Lake Malawi. CONCLUSION Alphavirus infections may contribute significantly to the febrile disease burden in the study area, and are associated with several arthropod-borne infections. Their spread seems only limited by factors affecting mosquitoes, and seems less restricted than that of Rift Valley fever.
Collapse
Affiliation(s)
- Nina Weller
- Division of Infectious Diseases and Tropical Medicine, Medical Centre of the University of Munich (LMU), Munich, Germany
| | - Petra Clowes
- National Institute for Medical Research -Mbeya Medical Research Center, Mbeya, Tanzania
| | - Gerhard Dobler
- Bundeswehr Institute of Microbiology, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Elmar Saathoff
- Division of Infectious Diseases and Tropical Medicine, Medical Centre of the University of Munich (LMU), Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Inge Kroidl
- Division of Infectious Diseases and Tropical Medicine, Medical Centre of the University of Munich (LMU), Munich, Germany
- National Institute for Medical Research -Mbeya Medical Research Center, Mbeya, Tanzania
| | | | - Leonard Maboko
- National Institute for Medical Research -Mbeya Medical Research Center, Mbeya, Tanzania
| | - Thomas Löscher
- Division of Infectious Diseases and Tropical Medicine, Medical Centre of the University of Munich (LMU), Munich, Germany
| | - Michael Hoelscher
- Division of Infectious Diseases and Tropical Medicine, Medical Centre of the University of Munich (LMU), Munich, Germany
- National Institute for Medical Research -Mbeya Medical Research Center, Mbeya, Tanzania
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Norbert Heinrich
- Division of Infectious Diseases and Tropical Medicine, Medical Centre of the University of Munich (LMU), Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
- * E-mail:
| |
Collapse
|
325
|
Montero A. [Chikungunya fever - A new global threat]. Med Clin (Barc) 2014; 145:118-23. [PMID: 25087211 PMCID: PMC7094327 DOI: 10.1016/j.medcli.2014.05.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 05/22/2014] [Accepted: 05/29/2014] [Indexed: 11/28/2022]
Abstract
Las epidemias causadas, entre otros, por los virus Ébola, Marburgo, Nipah, Lassa, coronavirus, virus del Nilo Occidental, virus de Saint Louis, virus de la inmunodeficiencia humana, dengue, fiebre amarilla y fiebre hemorrágica venezolana han puesto sobre el tapete el riesgo que estos agentes representan para la salud pública global. Entre las nuevas amenazas destaca el virus chikungunya, que ha extendido rápidamente su área endémica desde regiones remotas de África hacia la cuenca del océano Índico y el Pacífico Oriental, causando importantes epidemias en África, Asia, islas del Índico y Europa Occidental, llegando a establecerse recientemente en islas del Caribe. Debido a su comportamiento dual endémico y epidémico, a sus propiedades virológicas y a la presencia global de sus vectores, este virus entraña el riesgo de causar grandes epidemias e instalarse en el territorio continental de las Américas a partir de la introducción de casos importados y a la circulación local vista en la región caribeña. Se revisan las principales características epidemiológicas y clínicas de la fiebre chikungunya y el riesgo de introducción de esta enfermedad en las Américas, enfatizando el rol de la vigilancia y la lucha contra los mosquitos en su prevención.
Collapse
Affiliation(s)
- Antonio Montero
- Miembro de la Carrera del Investigador Científico, Consejo de Investigaciones de la Universidad Nacional de Rosario (CIUNR), Rosario, Santa Fe, Argentina; Centro de Medicina Tropical y Enfermedades Infecciosas Emergentes, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina.
| |
Collapse
|
326
|
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
|
327
|
Weger-Lucarelli J, Chu H, Aliota MT, Partidos CD, Osorio JE. A novel MVA vectored Chikungunya virus vaccine elicits protective immunity in mice. PLoS Negl Trop Dis 2014; 8:e2970. [PMID: 25058320 PMCID: PMC4109897 DOI: 10.1371/journal.pntd.0002970] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 05/13/2014] [Indexed: 12/20/2022] Open
Abstract
Background Chikungunya virus (CHIKV) is a re-emerging arbovirus associated with febrile illness often accompanied by rash and arthralgia that may persist for several years. Outbreaks are associated with high morbidity and create a public health challenge for countries affected. Recent outbreaks have occurred in both Europe and the Americas, suggesting CHIKV may continue to spread. Despite the sustained threat of the virus, there is no approved vaccine or antiviral therapy against CHIKV. Therefore, it is critical to develop a vaccine that is both well tolerated and highly protective. Methodology/Principal Findings In this study, we describe the construction and characterization of a modified Vaccinia virus Ankara (MVA) virus expressing CHIKV E3 and E2 proteins (MVA-CHIK) that protected several mouse models from challenge with CHIKV. In particular, BALB/c mice were completely protected against viremia upon challenge with CHIKV after two doses of MVA-CHIK. Additionally, A129 mice (deficient in IFNα/β) were protected from viremia, footpad swelling, and mortality. While high anti-virus antibodies were elicited, low or undetectable levels of neutralizing antibodies were produced in both mouse models. However, passive transfer of MVA-CHIK immune serum to naïve mice did not protect against mortality, suggesting that antibodies may not be the main effectors of protection afforded by MVA-CHIK. Furthermore, depletion of CD4+, but not CD8+ T-cells from vaccinated mice resulted in 100% mortality, implicating the indispensable role of CD4+ T-cells in the protection afforded by MVA-CHIK. Conclusions/Significance The results presented herein demonstrate the potential of MVA to effectively express CHIKV E3-E2 proteins and generate protective immune responses. Our findings challenge the assumption that only neutralizing antibodies are effective in providing protection against CHIKV, and provides a framework for the development of novel, more effective vaccine strategies to combat CHIKV. Chikungunya virus (CHIKV) has recently re-emerged from Africa to cause disease outbreaks in Asia, Europe, and more recently the Caribbean. The virus is transmitted by Aedes mosquitoes and causes a disease that is characterized by high fever and incapacitating joint pain that can cause great personal and economic loss. At present, no approved vaccine or antivirals are approved against CHIKV. In this study, we developed a novel CHIKV vaccine that is vectored by Modified Vaccinia virus Ankara (MVA), an attenuated vaccine vector which has been shown to be safe in humans and induce a strong immune response. The vaccine expresses the E3 and E2 proteins of CHIKV, the latter of which is thought to be the main mediator of protection. The vaccine was effective in two mouse models and protected against all markers of disease tested despite the absence of high levels of neutralizing antibodies, the gold standard of protection. Depletion of CD4+ T cells from vaccinated mice resulted in loss of protection, implicating these cells in the protection induced by the vaccine.
Collapse
Affiliation(s)
- James Weger-Lucarelli
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| | - Haiyan Chu
- Takeda, Inc., Madison, Wisconsin, United States of America
| | - Matthew T. Aliota
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | | | - Jorge E. Osorio
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
328
|
Mahauad-Fernandez WD, Jones PH, Okeoma CM. Critical role for bone marrow stromal antigen 2 in acute Chikungunya virus infection. J Gen Virol 2014; 95:2450-2461. [PMID: 25053563 DOI: 10.1099/vir.0.068643-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Bone marrow stromal antigen 2 (BST-2; also known as tetherin or CD317) is an IFN-inducible gene that functions to block the release of a range of nascent enveloped virions from infected host cells. However, the role of BST-2 in viral pathogenesis remains poorly understood. BST-2 plays a multifaceted role in innate immunity, as it hinders retroviral infection and possibly promotes infection with some rhabdo- and orthomyxoviruses. This paradoxical role has probably hindered exploration of BST-2 antiviral function in vivo. We reported previously that BST-2 tethers Chikungunya virus (CHIKV)-like particles on the cell plasma membrane. To explore the role of BST-2 in CHIKV replication and host protection, we utilized CHIKV strain 181/25 to examine early events during CHIKV infection in a BST-2(-/-) mouse model. We observed an interesting dichotomy between WT and BST-2(-/-) mice. BST-2 deficiency increased inoculation site viral load, culminating in higher systemic viraemia and increased lymphoid tissues tropism. A suppressed inflammatory innate response demonstrated by impaired expression of IFN-α, IFN-γ and CD40 ligand was observed in BST-2(-/-) mice compared with the WT controls. These findings suggested that, in part, BST-2 protects lymphoid tissues from CHIKV infection and regulates CHIKV-induced inflammatory response by the host.
Collapse
Affiliation(s)
- Wadie D Mahauad-Fernandez
- Interdisciplinary Graduate Program in Molecular and Cellular Biology (MCB), University of Iowa, Iowa City, IA, USA.,Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Philip H Jones
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Chioma M Okeoma
- Interdisciplinary Graduate Program in Molecular and Cellular Biology (MCB), University of Iowa, Iowa City, IA, USA.,Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
329
|
Bioactive cembrane derivatives from the Indian Ocean soft coral, Sinularia kavarattiensis. Mar Drugs 2014; 12:4045-68. [PMID: 25056629 PMCID: PMC4113814 DOI: 10.3390/md12074045] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/05/2014] [Accepted: 06/17/2014] [Indexed: 12/21/2022] Open
Abstract
Marine organisms and their metabolites represent a unique source of potential pharmaceutical substances. In this study, we examined marine-derived substances for their bioactive properties in a cell-based Chikungunya virus (CHIKV) replicon model and for in vitro anti-inflammatory activity. In the screening of a marine sample library, crude extracts from the Indian soft coral, Sinularia kavarattiensis, showed promising activity against the CHIKV replicon. Bioassay-guided chemical fractionation of S. kavarattiensis resulted in the isolation of six known norcembranoids (1-6) and one new compound, named kavaranolide (7). The structures were elucidated on the basis of NMR and MS spectroscopic data. Compounds 1-3 and 5-7 were evaluated for their replicon-inhibiting potential in the CHIKV model by using a luminescence-based detection technique and live cell imaging. Compounds 1 and 2 showed moderate inhibition of the CHIKV replicon, but imaging studies also revealed cytotoxic properties. Moreover, the effects of the isolated compounds on primary microglial cells, an experimental model for neuroinflammation, were evaluated. Compound 2 was shown to modulate the immune response in microglial cells and to possess potential anti-inflammatory properties by dose-dependently reducing the release of pro- and anti-inflammatory cytokines.
Collapse
|
330
|
Shytuhina A, Pristatsky P, He J, Casimiro DR, Schwartz RM, Hoang VM, Ha S. Development and application of a reversed-phase high-performance liquid chromatographic method for quantitation and characterization of a Chikungunya virus-like particle vaccine. J Chromatogr A 2014; 1364:192-7. [PMID: 25234500 DOI: 10.1016/j.chroma.2014.05.087] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/20/2014] [Accepted: 05/30/2014] [Indexed: 11/19/2022]
Abstract
To effectively support the development of a Chikungunya (CHIKV) virus-like particle (VLP) vaccine, a sensitive and robust high-performance liquid chromatography (HPLC) method that can quantitate CHIKV VLPs and monitor product purity throughout the manufacturing process is needed. We developed a sensitive reversed-phase HPLC (RP-HPLC) method that separates capsid, E1, and E2 proteins in CHIKV VLP vaccine with good resolution. Each protein component was verified by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and matrix-assisted laser desorption/ionization time-of-flight (MALDI-ToF) mass spectrometry (MS). The post-translational modifications on the viral glycoproteins E1 and E2 were further identified by intact protein mass measurements with liquid chromatography-mass spectrometry (LC-MS). The RP-HPLC method has a linear range of 0.51-12 μg protein, an accuracy of 96-106% and a precision of 12% RSD, suitable for vaccine product release testing. In addition, we demonstrated that the RP-HPLC method is useful for characterizing viral glycoprotein post-translational modifications, monitoring product purity during process development and assessing product stability during formulation development.
Collapse
Affiliation(s)
- Anastasija Shytuhina
- Vaccine Bioprocess Research & Development, Merck Research Laboratories, West Point, PA, 19486, United States
| | - Pavlo Pristatsky
- Vaccine Bioprocess Research & Development, Merck Research Laboratories, West Point, PA, 19486, United States
| | - Jian He
- Vaccine Bioprocess Research & Development, Merck Research Laboratories, West Point, PA, 19486, United States
| | - Danilo R Casimiro
- Vaccine Bioprocess Research & Development, Merck Research Laboratories, West Point, PA, 19486, United States
| | - Richard M Schwartz
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Gaithersburg, MD 20878, United States
| | - Van M Hoang
- Vaccine Bioprocess Research & Development, Merck Research Laboratories, West Point, PA, 19486, United States
| | - Sha Ha
- Vaccine Bioprocess Research & Development, Merck Research Laboratories, West Point, PA, 19486, United States.
| |
Collapse
|
331
|
Locking and blocking the viral landscape of an alphavirus with neutralizing antibodies. J Virol 2014; 88:9616-23. [PMID: 24920796 DOI: 10.1128/jvi.01286-14] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED Alphaviruses are serious, sometimes lethal human pathogens that belong to the family Togaviridae. The structures of human Venezuelan equine encephalitis virus (VEEV), an alphavirus, in complex with two strongly neutralizing antibody Fab fragments (F5 and 3B4C-4) have been determined using a combination of cryo-electron microscopy and homology modeling. We characterize these monoclonal antibody Fab fragments, which are known to abrogate VEEV infectivity by binding to the E2 (envelope) surface glycoprotein. Both of these antibody Fab fragments cross-link the surface E2 glycoproteins and therefore probably inhibit infectivity by blocking the conformational changes that are required for making the virus fusogenic. The F5 Fab fragment cross-links E2 proteins within one trimeric spike, whereas the 3B4C-4 Fab fragment cross-links E2 proteins from neighboring spikes. Furthermore, F5 probably blocks the receptor-binding site, whereas 3B4C-4 sterically hinders the exposure of the fusion loop at the end of the E2 B-domain. IMPORTANCE Alphaviral infections are transmitted mainly by mosquitoes. Venezuelan equine encephalitis virus (VEEV) is an alphavirus with a wide distribution across the globe. No effective vaccines exist for alphaviral infections. Therefore, a better understanding of VEEV and its associated neutralizing antibodies will help with the development of effective drugs and vaccines.
Collapse
|
332
|
Khan K, Bogoch I, Brownstein JS, Miniota J, Nicolucci A, Hu W, Nsoesie EO, Cetron M, Creatore MI, German M, Wilder-Smith A. Assessing the origin of and potential for international spread of chikungunya virus from the Caribbean. PLOS CURRENTS 2014; 6:ecurrents.outbreaks.2134a0a7bf37fd8d388181539fea2da5. [PMID: 24944846 PMCID: PMC4055609 DOI: 10.1371/currents.outbreaks.2134a0a7bf37fd8d388181539fea2da5] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND For the first time, an outbreak of chikungunya has been reported in the Americas. Locally acquired infections have been confirmed in fourteen Caribbean countries and dependent territories, Guyana and French Guiana, in which a large number of North American travelers vacation. Should some travelers become infected with chikungunya virus, they could potentially introduce it into the United States, where there are competent Aedes mosquito vectors, with the possibility of local transmission. METHODS We analyzed historical data on airline travelers departing areas of the Caribbean and South America, where locally acquired cases of chikungunya have been confirmed as of May 12th, 2014. The final destinations of travelers departing these areas between May and July 2012 were determined and overlaid on maps of the reported distribution of Aedes aeygpti and albopictus mosquitoes in the United States, to identify potential areas at risk of autochthonous transmission. RESULTS The United States alone accounted for 52.1% of the final destinations of all international travelers departing chikungunya indigenous areas of the Caribbean between May and July 2012. Cities in the United States with the highest volume of air travelers were New York City, Miami and San Juan (Puerto Rico). Miami and San Juan were high travel-volume cities where Aedes aeygpti or albopictus are reported and where climatic conditions could be suitable for autochthonous transmission. CONCLUSION The rapidly evolving outbreak of chikungunya in the Caribbean poses a growing risk to countries and areas linked by air travel, including the United States where competent Aedes mosquitoes exist. The risk of chikungunya importation into the United States may be elevated following key travel periods in the spring, when large numbers of North American travelers typically vacation in the Caribbean.
Collapse
Affiliation(s)
- Kamran Khan
- Department of Medicine, Division of Infectious Diseases, University of Toronto, Toronto, Canada
| | - Isaac Bogoch
- Department of Medicine, Division of Infectious Diseases, University of Toronto, Toronto, Canada; University Health Network, Divisions of Internal Medicine and Infectious Diseases, Toronto, Canada
| | - John S Brownstein
- Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer Miniota
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada
| | - Adrian Nicolucci
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada
| | - Wei Hu
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada
| | - Elaine O Nsoesie
- Children's Hospital Informatics Program, Boston Children's Hospital, Boston, Massachusetts, USA
; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA ; Network Dynamics and Simulation Science Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, USA
| | - Martin Cetron
- Division of Global Migration and Quarantine, Centers for Disease Control and Prevention, Atlanta, USA; Departments of Medicine and Epidemiology, Emory University School of Medicine and Rollins School of Public Health, Atlanta, USA
| | | | - Matthew German
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada
| | - Annelies Wilder-Smith
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Institute of Public Health, University of Heidelberg, Germany
| |
Collapse
|
333
|
Priya R, Patro IK, Parida MM. TLR3 mediated innate immune response in mice brain following infection with Chikungunya virus. Virus Res 2014; 189:194-205. [PMID: 24905288 DOI: 10.1016/j.virusres.2014.05.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 05/08/2014] [Accepted: 05/09/2014] [Indexed: 12/12/2022]
Abstract
Chikungunya virus (CHIKV) has received global attention due to the series of large-scale outbreaks in different parts of the world. Many unusual clinical severities including neurological complications and death were reported in recent outbreaks. The mechanism underlying the host immune response to CHIKV in the brain is poorly characterized. In this study, the neuropathogenesis of CHIKV with E1:A226V mutation was elucidated in 1 week old BALB/c mice. The virus was found to replicate in mice brain with peak titer of 10(4) on 6th day post infection. Immunohistochemical analysis revealed preferential virus localization in neuronal cells of cerebellum. The expression profiling of TLR, antiviral genes and cytokines in mice brain revealed significant up regulation of TLR3, TRAF-6, TICAM-1, MCP-1, CXCL-10, IL-6, IL-4, ISG-15, MX-2, IFN-β, OAS-3 genes that ultimately resulted in virus clearance from brain by day 9-10 suggesting activation of innate immune pathway. Further the effect of poly I: C (Polyinosinic: Polycytidylic acid), a TLR-3 agonist and potent IFN inducer on CHIKV neuropathogenesis was studied. Pretreatment of mice with Poly I: C caused reduction of CHIKV titer in brain and offered 100% protection of animals. The protection was mediated by an increased induction of TLR3, IFN-β and antiviral genes in mice brain. Our result demonstrates that pre immune stimulation of animals by Poly I: C is effective inhibitor of CHIKV replication and might be a promising prevention agent against this virus.
Collapse
Affiliation(s)
- Raj Priya
- Division of Virology, Defence Research & Development Establishment, Gwalior 474002, India
| | - I K Patro
- School of Studies in Neurosciences, Jiwaji University, Gwalior 474002, India
| | - M M Parida
- Division of Virology, Defence Research & Development Establishment, Gwalior 474002, India.
| |
Collapse
|
334
|
Stoler J, Al Dashti R, Anto F, Fobil JN, Awandare GA. Deconstructing "malaria": West Africa as the next front for dengue fever surveillance and control. Acta Trop 2014; 134:58-65. [PMID: 24613157 DOI: 10.1016/j.actatropica.2014.02.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 02/19/2014] [Accepted: 02/23/2014] [Indexed: 11/15/2022]
Abstract
Presumptive treatment of febrile illness patients for malaria remains the norm in endemic areas of West Africa, and "malaria" remains the top source of health facility outpatient visits in many West African nations. Many other febrile illnesses, including bacterial, viral, and fungal infections, share a similar symptomatology as malaria and are routinely misdiagnosed as such; yet growing evidence suggests that much of the burden of febrile illness is often not attributable to malaria. Dengue fever is one of several viral diseases with symptoms similar to malaria, and the combination of rapid globalization, the long-standing presence of Aedes mosquitoes, case reports from travelers, and recent seroprevalence surveys all implicate West Africa as an emerging front for dengue surveillance and control. This paper integrates recent vector ecology, public health, and clinical medicine literature about dengue in West Africa across community, regional, and global geographic scales. We present a holistic argument for greater attention to dengue fever surveillance in West Africa and renew the call for improving differential diagnosis of febrile illness patients in the region.
Collapse
Affiliation(s)
- Justin Stoler
- Department of Geography and Regional Studies, University of Miami, 1300 Campo Sano Avenue, Coral Gables, FL, USA; Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - Rawan Al Dashti
- Department of Public Health Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - Francis Anto
- Department of Epidemiology and Disease Control, University of Ghana, Legon, Ghana.
| | - Julius N Fobil
- Department of Biological, Environmental & Occupational Health Sciences, University of Ghana, Legon, Ghana.
| | - Gordon A Awandare
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Ghana.
| |
Collapse
|
335
|
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
|
336
|
Paingankar MS, Arankalle VA. Identification of chikungunya virus interacting proteins in mammalian cells. J Biosci 2014; 39:389-99. [DOI: 10.1007/s12038-014-9436-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
337
|
Discovery of in silico hits targeting the nsP3 macro domain of chikungunya virus. J Mol Model 2014; 20:2216. [PMID: 24756552 PMCID: PMC7088235 DOI: 10.1007/s00894-014-2216-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 03/16/2014] [Indexed: 12/24/2022]
Abstract
The recent emergence and re-emergence of alphaviruses, in particular the chikungunya virus (CHIKV), in numerous countries has invoked a worldwide threat to human health, while simultaneously generating an economic burden on affected countries. There are currently no vaccines or effective drugs available for the treatment of the CHIKV, and with few lead compounds reported, the vital medicinal chemistry is significantly more challenging. This study reports on the discovery of potential inhibitors for the nsP3 macro domain of CHIKV using molecular docking, virtual screening, and molecular dynamics simulations, as well as work done to evaluate and confirm the active site of nsP3. Virtual screening was carried out based on blind docking as well as focused docking, using the database of 1541 compounds from NCI Diversity Set II, to identify hit compounds for nsP3. The top hit compounds were further subjected to molecular dynamic simulations, yielding a greater understanding of the dynamic behavior of nsP3 and its complexes with various ligands, concurrently confirming the outcomes of docking, and establishing in silico lead compounds which target the CHIKV nsP3 enzyme. Virtual screening identifies novel inhibitors targeting the nsP3 macro domain of chikungunya virus ![]()
Collapse
|
338
|
Kam YW, Lee WWL, Simarmata D, Le Grand R, Tolou H, Merits A, Roques P, Ng LFP. Unique epitopes recognized by antibodies induced in Chikungunya virus-infected non-human primates: implications for the study of immunopathology and vaccine development. PLoS One 2014; 9:e95647. [PMID: 24755730 PMCID: PMC3995782 DOI: 10.1371/journal.pone.0095647] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 03/28/2014] [Indexed: 11/17/2022] Open
Abstract
Chikungunya virus (CHIKV) is an Alphavirus that causes chronic and incapacitating arthralgia in humans. Although patient cohort studies have shown the production of CHIKV specific antibodies, the fine specificity of the antibody response against CHIKV is not completely defined. The macaque model of CHIKV infection was established due to limitations of clinical specimens. More importantly, its close relation to humans will allow the study of chronic infection and further identify important CHIKV targets. In this study, serum samples from CHIKV-infected macaques collected at different time-points post infection were used to characterize the antibody production pattern and kinetics. Results revealed that anti-CHIKV antibodies were neutralizing and the E2 glycoprotein, Capsid, nsP1, nsP3 and nsP4 proteins were targets of the anti-CHIKV antibody response in macaques. Furthermore, linear B-cell epitopes recognized by these anti-CHIKV antibodies were identified, and mapped to their structural localization. This characterizes the specificity of anti-CHIKV antibody response in macaques and further demonstrates the importance of the different regions in CHIKV-encoded proteins in the adaptive immune response. Information from this study provides critical knowledge that will aid in the understanding of CHIKV infection and immunity, vaccine design, and pre-clinical efficacy studies.
Collapse
Affiliation(s)
- Yiu-Wing Kam
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Wendy W L Lee
- Singapore Immunology Network, 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
| | - Diane Simarmata
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Roger Le Grand
- CEA, Division of ImmunoVirology (SIV), Institute of Emerging Diseases and Innovative Therapies (IMETI), Fontenay-aux-Roses, France; Université Paris-Sud 11, UMR E1, Orsay, France
| | - Hugues Tolou
- Groupe d'Etude en Préventologie (GEP), Villenave d'Ornon, France
| | - Andres Merits
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Pierre Roques
- CEA, Division of ImmunoVirology (SIV), Institute of Emerging Diseases and Innovative Therapies (IMETI), Fontenay-aux-Roses, France; Université Paris-Sud 11, UMR E1, Orsay, France
| | - Lisa F P Ng
- Singapore Immunology Network, 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
| |
Collapse
|
339
|
Wadhwani GG. Homeopathic drug therapy. Homeopathy in Chikungunya Fever and Post-Chikungunya Chronic Arthritis: an observational study. HOMEOPATHY 2014; 102:193-8. [PMID: 23870379 DOI: 10.1016/j.homp.2013.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 01/18/2013] [Accepted: 02/05/2013] [Indexed: 11/18/2022]
Abstract
OBJECTIVES To observe the effect of homeopathic therapy in Chikungunya Fever (CF) and in Post-Chikungunya Chronic Arthritis (PCCA) in a primary health care setting. METHODS A prospective observational study was conducted at Delhi Government Homeopathic Dispensary, Aali Village, New Delhi, India, for a period of 6 months, from 1st October 2010 to 31st March 2011. 126 patients (75 CF, 51 PCCA) were enrolled based on predefined inclusion criteria. A single homeopathic medicine was prescribed for each patient after case taking with the help of Materia Medica and/or Repertory. Results were evaluated on the basis of visual analogue scale and symptom scores. RESULTS Complete recovery was seen in 84.5% CF cases in a mean time of 6.8 days. 90% cases of PCCA recovered completely in a mean time of 32.5 days. CONCLUSION Homeopathic therapy may be effective in CF and PCCA. A randomized controlled trial should be considered.
Collapse
Affiliation(s)
- Gyandas G Wadhwani
- Directorate of ISM & Homeopathy, Govt of National Capital Territory of Delhi, India.
| |
Collapse
|
340
|
Olagnier D, Scholte FEM, Chiang C, Albulescu IC, Nichols C, He Z, Lin R, Snijder EJ, van Hemert MJ, Hiscott J. Inhibition of dengue and chikungunya virus infections by RIG-I-mediated type I interferon-independent stimulation of the innate antiviral response. J Virol 2014; 88:4180-94. [PMID: 24478443 PMCID: PMC3993760 DOI: 10.1128/jvi.03114-13] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 01/20/2014] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED RIG-I is a cytosolic sensor critically involved in the activation of the innate immune response to RNA virus infection. In the present study, we evaluated the inhibitory effect of a RIG-I agonist on the replication of two emerging arthropod-borne viral pathogens, dengue virus (DENV) and chikungunya virus (CHIKV), for which no therapeutic options currently exist. We demonstrate that when a low, noncytotoxic dose of an optimized 5'triphosphorylated RNA (5'pppRNA) molecule was administered, RIG-I stimulation generated a robust antiviral response against these two viruses. Strikingly, 5'pppRNA treatment before or after challenge with DENV or CHIKV provided protection against infection. In primary human monocytes and monocyte-derived dendritic cells, the RIG-I agonist blocked both primary infection and antibody-dependent enhancement of DENV infection. The protective response against DENV and CHIKV induced by 5'pppRNA was dependent on an intact RIG-I/MAVS/TBK1/IRF3 axis and was largely independent of the type I IFN response. Altogether, this in vitro analysis of the antiviral efficacy of 5'pppRNA highlights the therapeutic potential of RIG-I agonists against emerging viruses such as DENV and CHIKV. IMPORTANCE DENV and CHIKV are two reemerging mosquito-borne viruses for which no therapeutic options currently exist. Both viruses overlap geographically in tropical regions of the world, produce similar fever-like symptoms, and are difficult to diagnose. This study investigated the inhibitory effect of a RIG-I agonist on the replication of these two viruses. RIG-I stimulation using 5'pppRNA before or after DENV or CHIKV infection generated a protective antiviral response against both pathogens in immune and nonimmune cells; interestingly, the protective response against the viruses was largely independent of the classical type I interferon response. The antiviral efficacy of 5'pppRNA highlights the therapeutic potential of RIG-I agonists against emerging viruses such as DENV and CHIKV.
Collapse
Affiliation(s)
- David Olagnier
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Florine E. M. Scholte
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cindy Chiang
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Irina C. Albulescu
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Carmen Nichols
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Zhong He
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| | - Rongtuan Lin
- Lady Davis Institute, Jewish General Hospital–McGill University, Montreal, Canada
| | - Eric J. Snijder
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Martijn J. van Hemert
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - John Hiscott
- Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida, USA
| |
Collapse
|
341
|
High level of vector competence of Aedes aegypti and Aedes albopictus from ten American countries as a crucial factor in the spread of Chikungunya virus. J Virol 2014; 88:6294-306. [PMID: 24672026 DOI: 10.1128/jvi.00370-14] [Citation(s) in RCA: 286] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
UNLABELLED Chikungunya virus (CHIKV) causes a major public health problem. In 2004, CHIKV began an unprecedented global expansion and has been responsible for epidemics in Africa, Asia, islands in the Indian Ocean region, and surprisingly, in temperate regions, such as Europe. Intriguingly, no local transmission of chikungunya virus (CHIKV) had been reported in the Americas until recently, despite the presence of vectors and annually reported imported cases. Here, we assessed the vector competence of 35 American Aedes aegypti and Aedes albopictus mosquito populations for three CHIKV genotypes. We also compared the number of viral particles of different CHIKV strains in mosquito saliva at two different times postinfection. Primarily, viral dissemination rates were high for all mosquito populations irrespective of the tested CHIKV isolate. In contrast, differences in transmission efficiency (TE) were underlined in populations of both species through the Americas, suggesting the role of salivary glands in selecting CHIKV for highly efficient transmission. Nonetheless, both mosquito species were capable of transmitting all three CHIKV genotypes, and TE reached alarming rates as high as 83.3% and 96.7% in A. aegypti and A. albopictus populations, respectively. A. albopictus better transmitted the epidemic mutant strain CHIKV_0621 of the East-Central-South African (ECSA) genotype than did A. aegypti, whereas the latter species was more capable of transmitting the original ECSA CHIKV_115 strain and also the Asian genotype CHIKV_NC. Therefore, a high risk of establishment and spread of CHIKV throughout the tropical, subtropical, and even temperate regions of the Americas is more real than ever. IMPORTANCE Until recently, the Americas had never reported chikungunya (CHIK) autochthonous transmission despite its global expansion beginning in 2004. Large regions of the continent are highly infested with Aedes aegypti and Aedes albopictus mosquitoes, and millions of dengue (DEN) cases are annually recorded. Indeed, DEN virus and CHIK virus (CHIKV) share the same vectors. Due to a recent CHIK outbreak affecting Caribbean islands, the need for a Pan-American evaluation of vector competence was compelling as a key parameter in assessing the epidemic risk. We demonstrated for the first time that A. aegypti and A. albopictus populations throughout the continent are highly competent to transmit CHIK irrespective of the viral genotypes tested. The risk of CHIK spreading throughout the tropical, subtropical, and even temperate regions of the Americas is more than ever a reality. In light of our results, local authorities should immediately pursue and reinforce epidemiological and entomological surveillance to avoid a severe epidemic.
Collapse
|
342
|
Arias-Goeta C, Moutailler S, Mousson L, Zouache K, Thiberge JM, Caro V, Rougeon F, Failloux AB. Chikungunya virus adaptation to a mosquito vector correlates with only few point mutations in the viral envelope glycoprotein. INFECTION GENETICS AND EVOLUTION 2014; 24:116-26. [PMID: 24681263 DOI: 10.1016/j.meegid.2014.03.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 02/27/2014] [Accepted: 03/17/2014] [Indexed: 01/06/2023]
Abstract
Like most arthropod-borne viruses (arboviruses), chikungunya virus (CHIKV) is a RNA virus maintained in nature in an alternating cycle of replication between invertebrate and vertebrate hosts. It has been assumed that host alternation restricts arbovirus genome evolution and imposes fitness trade-offs. Despite their slower rates of evolution, arboviruses still have the capacity to produce variants capable to exploit new environments. To test whether the evolution of the newly emerged epidemic variant of CHIKV (E1-226V) is constrained by host alternation, the virus was alternately-passaged in hamster-derived BHK-21 cells and Aedes aegypti-derived Aag-2 cells. It was also serially-passaged in BHK-21 or Aag-2 cells to promote adaptation to one cell type and presumably, fitness cost in the bypassed cell type. After 30 passages, obtained CHIKV strains were genetically and phenotypically characterized using in vitro and in vivo systems. Serially- and alternately-passaged strains can be distinguished by amino-acid substitutions in the E2 glycoprotein, responsible for receptor binding. Two substitutions at positions E2-64 and E2-208 only lower the dissemination of the variant E1-226V in Ae. aegypti. These amino-acid changes in the E2 glycoprotein might affect viral infectivity by altering the interaction between CHIKV E1-226V and the cellular receptor on the midgut epithelial cells in Ae. aegypti but not in Aedesalbopictus.
Collapse
Affiliation(s)
- Camilo Arias-Goeta
- Department of Virology, Arboviruses and Insect Vectors, Institut Pasteur, Paris, France; Cellule Pasteur UPMC, Université Pierre et Marie Curie, Paris, France.
| | - Sara Moutailler
- Department of Virology, Arboviruses and Insect Vectors, Institut Pasteur, Paris, France
| | - Laurence Mousson
- Department of Virology, Arboviruses and Insect Vectors, Institut Pasteur, Paris, France
| | - Karima Zouache
- Department of Virology, Arboviruses and Insect Vectors, Institut Pasteur, Paris, France
| | - Jean-Michel Thiberge
- Department of Infection and Epidemiology, Genotyping of Pathogens and Public Health, Institut Pasteur, Paris, France
| | - Valérie Caro
- Department of Infection and Epidemiology, Genotyping of Pathogens and Public Health, Institut Pasteur, Paris, France
| | - François Rougeon
- URA 2581, Genetic and Molecular Interactions Cell-eucaryote, Institut Pasteur, Paris, France
| | - Anna-Bella Failloux
- Department of Virology, Arboviruses and Insect Vectors, Institut Pasteur, Paris, France
| |
Collapse
|
343
|
Abstract
Virus infections usually begin in peripheral tissues and can invade the mammalian nervous system (NS), spreading into the peripheral (PNS) and more rarely the central (CNS) nervous systems. The CNS is protected from most virus infections by effective immune responses and multilayer barriers. However, some viruses enter the NS with high efficiency via the bloodstream or by directly infecting nerves that innervate peripheral tissues, resulting in debilitating direct and immune-mediated pathology. Most viruses in the NS are opportunistic or accidental pathogens, but a few, most notably the alpha herpesviruses and rabies virus, have evolved to enter the NS efficiently and exploit neuronal cell biology. Remarkably, the alpha herpesviruses can establish quiescent infections in the PNS, with rare but often fatal CNS pathology. Here we review how viruses gain access to and spread in the well-protected CNS, with particular emphasis on alpha herpesviruses, which establish and maintain persistent NS infections.
Collapse
Affiliation(s)
- Orkide O Koyuncu
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | | | | |
Collapse
|
344
|
Butt AM, Nasrullah I, Tong Y. Genome-wide analysis of codon usage and influencing factors in chikungunya viruses. PLoS One 2014; 9:e90905. [PMID: 24595095 PMCID: PMC3942501 DOI: 10.1371/journal.pone.0090905] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 02/06/2014] [Indexed: 02/03/2023] Open
Abstract
Chikungunya virus (CHIKV) is an arthropod-borne virus of the family Togaviridae that is transmitted to humans by Aedes spp. mosquitoes. Its genome comprises a 12 kb single-strand positive-sense RNA. In the present study, we report the patterns of synonymous codon usage in 141 CHIKV genomes by calculating several codon usage indices and applying multivariate statistical methods. Relative synonymous codon usage (RSCU) analysis showed that the preferred synonymous codons were G/C and A-ended. A comparative analysis of RSCU between CHIKV and its hosts showed that codon usage patterns of CHIKV are a mixture of coincidence and antagonism. Similarity index analysis showed that the overall codon usage patterns of CHIKV have been strongly influenced by Pan troglodytes and Aedes albopictus during evolution. The overall codon usage bias was low in CHIKV genomes, as inferred from the analysis of effective number of codons (ENC) and codon adaptation index (CAI). Our data suggested that although mutation pressure dominates codon usage in CHIKV, patterns of codon usage in CHIKV are also under the influence of natural selection from its hosts and geography. To the best of our knowledge, this is first report describing codon usage analysis in CHIKV genomes. The findings from this study are expected to increase our understanding of factors involved in viral evolution, and fitness towards hosts and the environment.
Collapse
Affiliation(s)
- Azeem Mehmood Butt
- Centre of Excellence in Molecular Biology (CEMB), University of the Punjab, Lahore, Pakistan
| | - Izza Nasrullah
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Yigang Tong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, People’s Republic of China
- * E-mail:
| |
Collapse
|
345
|
Lim PJ, Chu JJH. A polarized cell model for Chikungunya virus infection: entry and egress of virus occurs at the apical domain of polarized cells. PLoS Negl Trop Dis 2014; 8:e2661. [PMID: 24587455 PMCID: PMC3930524 DOI: 10.1371/journal.pntd.0002661] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 12/09/2013] [Indexed: 11/19/2022] Open
Abstract
Chikungunya virus (CHIKV) has resulted in several outbreaks in the past six decades. The clinical symptoms of Chikungunya infection include fever, skin rash, arthralgia, and an increasing incidence of encephalitis. The re-emergence of CHIKV with more severe pathogenesis highlights its potential threat on our human health. In this study, polarized HBMEC, polarized Vero C1008 and non-polarized Vero cells grown on cell culture inserts were infected with CHIKV apically or basolaterally. Plaque assays, viral binding assays and immunofluorescence assays demonstrated apical entry and release of CHIKV in polarized HBMEC and Vero C1008. Drug treatment studies were performed to elucidate both host cell and viral factors involved in the sorting and release of CHIKV at the apical domain of polarized cells. Disruption of host cell myosin II, microtubule and microfilament networks did not disrupt the polarized release of CHIKV. However, treatment with tunicamycin resulted in a bi-directional release of CHIKV, suggesting that N-glycans of CHIKV envelope glycoproteins could serve as apical sorting signals. Polarized cells are found in many parts of the human body and are characterized by the presence of two distinct plasma membrane domains: the apical domain facing the lumen and the basolateral domain facing the underlying tissues. Polarized epithelial cells line the major cavities of our body, while polarized endothelial cells line the blood-tissue interface, both of which protect our body against the invasion of biological pathogens. Thus, many pathogens have to invade the monolayer of epithelial or endothelial cells in order to establish infection. During infection with Chikungunya virus, a mosquito vector bites a human host and inoculates the virus into the host's bloodstream. In recent epidemics of Chikungunya infection, more severe clinical manifestations such as neurological complications were observed. As such, we studied the infection of Chikungunya virus in polarized cells in an aim to provide explanations for the more severe pathogenesis observed.
Collapse
Affiliation(s)
- Pei Jin Lim
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore, Singapore
| | - Justin Jang Hann Chu
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
346
|
Chusri S, Siripaitoon P, Silpapojakul K, Hortiwakul T, Charernmak B, Chinnawirotpisan P, Nisalak A, Thaisomboonsuk B, Klungthong C, Gibbons RV, Jarman RG. Kinetics of chikungunya infections during an outbreak in Southern Thailand, 2008-2009. Am J Trop Med Hyg 2014; 90:410-417. [PMID: 24493674 PMCID: PMC3945684 DOI: 10.4269/ajtmh.12-0681] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The Indian Ocean chikungunya epidemic re-emerged in Thailand in August 2008. Forty-five adults with laboratory-confirmed chikungunya in Songkhla province, Thailand were clinically assessed and serially bled throughout the acute and convalescent phase of the disease. Patient symptoms, antibody responses, and viral kinetics were evaluated using observational assessments, polymerase chain reaction (PCR), and serological assays. All subjects experienced joint pain with 42 (93%) involving multiple joints; the interphalangeal most commonly affected in 91% of the subjects. The mean duration of joint pain was 5.8 days, 11 (25%) experiencing discomfort through the duration of the study. Rash was observed in 37 (82%) subjects a mean 3.5 days post onset of symptoms. Patents were positive by PCR for a mean of 5.9 days with sustained peak viral load through Day 5. The IgM antibodies appeared on Day 4 and peaked at Day 7 and IgG antibodies first appeared at Day 5 and rose steadily through Day 24.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Richard G. Jarman
- *Address correspondence to Richard G. Jarman, Walter Reed Army Institute of Research, Viral Diseases Branch, 503 Robert Grant Avenue, Silver Spring, MD 20910. E-mail:
| |
Collapse
|
347
|
Rozen-Gagnon K, Stapleford KA, Mongelli V, Blanc H, Failloux AB, Saleh MC, Vignuzzi M. Alphavirus mutator variants present host-specific defects and attenuation in mammalian and insect models. PLoS Pathog 2014; 10:e1003877. [PMID: 24453971 PMCID: PMC3894214 DOI: 10.1371/journal.ppat.1003877] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 11/25/2013] [Indexed: 01/26/2023] Open
Abstract
Arboviruses cycle through both vertebrates and invertebrates, which requires them to adapt to disparate hosts while maintaining genetic integrity during genome replication. To study the genetic mechanisms and determinants of these processes, we use chikungunya virus (CHIKV), a re-emerging human pathogen transmitted by the Aedes mosquito. We previously isolated a high fidelity (or antimutator) polymerase variant, C483Y, which had decreased fitness in both mammalian and mosquito hosts, suggesting this residue may be a key molecular determinant. To further investigate effects of position 483 on RNA-dependent RNA-polymerase (RdRp) fidelity, we substituted every amino acid at this position. We isolated novel mutators with decreased replication fidelity and higher mutation frequencies, allowing us to examine the fitness of error-prone arbovirus variants. Although CHIKV mutators displayed no major replication defects in mammalian cell culture, they had reduced specific infectivity and were attenuated in vivo. Unexpectedly, mutator phenotypes were suppressed in mosquito cells and the variants exhibited significant defects in RNA synthesis. Consequently, these replication defects resulted in strong selection for reversion during infection of mosquitoes. Since residue 483 is conserved among alphaviruses, we examined the analogous mutations in Sindbis virus (SINV), which also reduced polymerase fidelity and generated replication defects in mosquito cells. However, replication defects were mosquito cell-specific and were not observed in Drosophila S2 cells, allowing us to evaluate the potential attenuation of mutators in insect models where pressure for reversion was absent. Indeed, the SINV mutator variant was attenuated in fruit flies. These findings confirm that residue 483 is a determinant regulating alphavirus polymerase fidelity and demonstrate proof of principle that arboviruses can be attenuated in mammalian and insect hosts by reducing fidelity. Chikungunya (CHIKV) is a re-emerging mosquito-borne virus that constitutes a major and growing human health burden. Like all RNA viruses, during viral replication CHIKV copies its genome using a polymerase that makes an average of one mistake per replication cycle. Therefore, a single virus generates millions of viral progeny that carry a multitude of distinct mutations in their genomes. In this study, we isolated CHIKV mutators (strains that make more errors than the wildtype virus), to study how higher mutation rates affect fitness in arthropod-borne viruses (arboviruses). CHIKV mutators have reduced virulence in mice and severe replication defects in Aedes mosquito cells. However, these replication defects result in selective pressure for reversion of mutators to a wildtype polymerase in mosquito hosts. To examine how mutators would behave in an insect model in absence of this genetic instability, we isolated mutators of a related virus, Sindbis virus (SINV). SINV mutators had no replication defect in fruit fly (Drosophila) cells, and a SINV mutator strain was stable and attenuated in fruit flies. This work shows proof of principle that arbovirus mutators can exhibit attenuation in both mammalian and insect hosts, and may remain a viable vaccine strategy.
Collapse
Affiliation(s)
- Kathryn Rozen-Gagnon
- Institut Pasteur, Viral Populations and Pathogenesis, CNRS UMR 3569, Paris, France
- University Paris Diderot, Sorbonne Paris Cite, Cellule Pasteur, Paris, France
| | | | - Vanesa Mongelli
- Institut Pasteur, Viruses and RNA Interference, UMR 3569, Paris, France
| | - Hervé Blanc
- Institut Pasteur, Viral Populations and Pathogenesis, CNRS UMR 3569, Paris, France
| | | | - Maria-Carla Saleh
- Institut Pasteur, Viruses and RNA Interference, UMR 3569, Paris, France
| | - Marco Vignuzzi
- Institut Pasteur, Viral Populations and Pathogenesis, CNRS UMR 3569, Paris, France
- * E-mail:
| |
Collapse
|
348
|
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
|
349
|
Roy CJ, Adams AP, Wang E, Plante K, Gorchakov R, Seymour RL, Vinet-Oliphant H, Weaver SC. Chikungunya vaccine candidate is highly attenuated and protects nonhuman primates against telemetrically monitored disease following a single dose. J Infect Dis 2014; 209:1891-9. [PMID: 24403555 DOI: 10.1093/infdis/jiu014] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that causes major epidemics of rash, fever, and debilitating arthritis. Currently, there are no vaccines or antivirals available for prevention or treatment. We therefore generated 2 live-attenuated vaccine candidates based on the insertion of a picornavirus internal ribosome entry site (IRES) sequence into the genome of CHIKV. Vaccination of cynomolgus macaques with a single dose of either vaccine produced no signs of disease but was highly immunogenic. After challenge with a subcutaneous inoculation of wild-type CHIKV, both vaccine candidates prevented the development of detectable viremia. Protected animals also exhibited no significant changes in core body temperature or cardiovascular rhythm, whereas sham-vaccinated animals showed hyperthermia, followed by sustained hypothermia, as well as significant changes in heart rate. These CHIKV/IRES vaccine candidates appear to be safe and efficacious, supporting their strong potential as human vaccines to protect against CHIKV infection and reduce transmission and further spread.
Collapse
Affiliation(s)
- Chad J Roy
- Division of Microbiology, Tulane National Primate Research Center, Covington, Louisiana
| | - A Paige Adams
- Institute for Human Infections and Immunity, Sealy Center for Vaccine Development Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| | - Eryu Wang
- Institute for Human Infections and Immunity, Sealy Center for Vaccine Development Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| | - Kenneth Plante
- Institute for Human Infections and Immunity, Sealy Center for Vaccine Development Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| | - Rodion Gorchakov
- Institute for Human Infections and Immunity, Sealy Center for Vaccine Development Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| | - Robert L Seymour
- Institute for Human Infections and Immunity, Sealy Center for Vaccine Development Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| | - Heather Vinet-Oliphant
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, Louisiana
| | - Scott C Weaver
- Institute for Human Infections and Immunity, Sealy Center for Vaccine Development Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
350
|
Evaluation of in vitro antiviral activity of Vitex Negundo L., Hyptis suaveolens (L) poit., Decalepis hamiltonii Wight & Arn., to Chikungunya virus. ASIAN PACIFIC JOURNAL OF TROPICAL DISEASE 2014. [DOI: 10.1016/s2222-1808(14)60424-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|