1
|
Gonçalves WA, de Sousa CDF, Teixeira MM, Souza DG. A brief overview of chikungunya-related pain. Eur J Pharmacol 2025; 994:177322. [PMID: 39892450 DOI: 10.1016/j.ejphar.2025.177322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 01/15/2025] [Accepted: 01/29/2025] [Indexed: 02/03/2025]
Abstract
Pain is an important symptom associated with the arboviral disease caused by the Chikungunya virus (CHIKV). For a significant number of patients, this symptom can persist for months or even years, negatively affecting their quality of life. Unfortunately, pharmacological options for this condition are limited and only partially effective, as the underlying mechanisms associated with CHIKV-induced pain are still poorly understood. The re-emergence of CHIKV has led to new outbreaks, and the expected high prevalence of pain in these global events requires new scientific advances to find more effective solutions. Here we review the main aspects of pain caused by CHIKV infection, such as the anatomy of the affected sites, the prevalence and management of this symptom, the diversity of possible cellular and molecular mechanisms, and finally highlight a promising meningeal pathway to elucidate the mechanisms involved in the unsolved problem of CHIKV-associated pain.
Collapse
Affiliation(s)
- William Antonio Gonçalves
- Departamento de Morfologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.
| | - Carla Daiane Ferreira de Sousa
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, University of Münster, Münster, Germany.
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.
| | - Daniele G Souza
- Laboratório Interação Microrganismo Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.
| |
Collapse
|
2
|
Cheong DHJ, Yi B, Wong YH, Chu JJH. The Current Progress in the Quest for Vaccines Against the Semliki Forest Virus Complex. Med Res Rev 2025; 45:947-967. [PMID: 39757142 DOI: 10.1002/med.22097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 10/10/2024] [Accepted: 12/12/2024] [Indexed: 01/07/2025]
Abstract
The Semliki Forest virus (SFV) complex comprises of arboviruses that are transmitted by arthropod vectors and cause acute febrile illness in humans. In the last seven decades, re-emergence of these viruses has resulted in numerous outbreaks globally, affecting regions including Africa, Americas, Asia, Europe and the Caribbean. These viruses are transmitted to humans by the bite of infected mosquitoes. Symptoms of infection include high fever, severe joint pain, skin rash, muscle pain and headache. Fatal cases were reported, and mortality rate increased during the epidemic of these viruses. There is therefore a need to control the spread of these emerging arboviruses. Given that vaccination is one of the most effective ways to protect populations against viral outbreaks, efforts have been made to develop and test potential vaccine candidates. However, there are still no licensed vaccines available against the medically important viruses in the SFV complex. This review first summarizes the current knowledge of the SFV complex disease pathogenesis. Next, seven strategies that have been applied in vaccine development against these viruses are reviewed, indicating the immune response and efficacies of these vaccine candidates in in vivo models of infection. Finally, the more promising candidates that have entered clinical trials are discussed and insights into the future development of vaccines for viruses of the SFV complex are given.
Collapse
Affiliation(s)
- Dorothy Hui Juan Cheong
- Department of Microbiology and Immunology, Infectious Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Bowen Yi
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Yi Hao Wong
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Justin Jang Hann Chu
- Department of Microbiology and Immunology, Infectious Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
- Collaborative and Translation Unit for Hand, Foot and Mouth Disease (HFMD), Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore CIty, Singapore
| |
Collapse
|
3
|
Piccinno R, Fiorenza G, Vasquez MI, Bouyer J, Notarides G, Gomulski LM, Meletiou S, Akiner M, Michaelakis A, Forneris F, Maga G, Gasperi G, Malacrida AR. On the tracks of an uninvited guest, the Asian tiger mosquito, Aedes albopictus in Cyprus. Parasit Vectors 2025; 18:39. [PMID: 39905537 PMCID: PMC11796127 DOI: 10.1186/s13071-024-06651-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/27/2024] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Aedes albopictus, the Asian tiger mosquito, which is listed among the world's 100 most dangerous invasive species, is the main vector of chikungunya, dengue and Zika viruses. This mosquito species has rapidly dispersed and invaded much of the globe assisted by its life history traits and high propagule pressure driven by human activities. Aedes albopictus is currently widespread across mainland Europe and the Mediterranean region, including the islands. Cyprus remained free of Ae. albopictus until October 2022, when specimens were recorded for the first time in Limassol district, including the port area. Understanding the processes associated with the introduction, expansion and establishment of this vector in Cyprus is of primary importance to mitigate its dispersal on the island, and to implement control methods to prevent disease outbreaks. A genetic analysis of these invasive specimens collected in Limassol district and in areas from the Central Mediterranean was performed to obtain a genetic portrait of the demographic history of the invasive mosquitoes on Cyprus. METHODS We applied highly polymorphic simple sequence repeat (SSR) markers to the Ae. albopictus mosquitoes collected in Cyprus and to specimens from Italy, France, Switzerland, the Balkans, Greece and Turkey to construct an SSR individual genotype dataset that would enable the invasion pattern of Ae. albopictus in Cyprus to be traced. Bayesian clustering analyses using STRUCTURE and BayesAss version 3 were employed to derive information on the degree of ancestry among Cypriot and Mediterranean mosquitoes and on recent mosquito movements both within Cyprus and between Cyprus and the Central Mediterranean areas. RESULTS The Cypriot mosquitoes appear to be highly polymorphic with no signs of genetic drift due to recent founder effects. An ongoing mosquito dispersal within the Limassol district was detected, suggesting the presence of established, hidden adventive populations. These mosquitoes share a high degree of ancestry with those in the Balkans and parts of northern Italy that border the Adriatic Sea. CONCLUSIONS Considering the trade connections of Limassol port, Cyprus with the Balkans and the Adriatic Italian region, we hypothesise that these areas may be involved in the incursion of Ae. albopictus into Cyprus. As the Balkan and Italian mosquitoes display high competence for CHIKV, questions arise about possible arbovirus outbreaks in Cyprus and highlight the need to implement surveillance and control measures.
Collapse
Affiliation(s)
- Riccardo Piccinno
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Giulia Fiorenza
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Marlen Ines Vasquez
- Department of Chemical Engineering, Cyprus University of Technology, Limassol, Cyprus
| | - Jeremy Bouyer
- Insect Pest Control Subprogramme, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency (IAEA), Vienna, Austria
- ASTRE, CIRAD, INRAE-University of Montpellier, Plateforme Technologique CYROI, Sainte-Clotilde, La Réunion, France
| | - Gregoris Notarides
- Department of Chemical Engineering, Cyprus University of Technology, Limassol, Cyprus
| | | | - Soteris Meletiou
- Department of Chemical Engineering, Cyprus University of Technology, Limassol, Cyprus
| | | | - Antonios Michaelakis
- Laboratory of Insects & Parasites of Medical Importance, Benaki Phytopathological Institute, Athens, Greece
| | - Federico Forneris
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Giovanni Maga
- Institute of Molecular Genetics of the National Research Council (IGM-CNR), Pavia, Italy
| | - Giuliano Gasperi
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy.
- Institute of Molecular Genetics of the National Research Council (IGM-CNR), Pavia, Italy.
| | | |
Collapse
|
4
|
Santiago-Cruz JA, Posadas-Mondragón A, Pérez-Juárez A, Herrera-González NE, Chin-Chan JM, Aguilar-González JE, Aguilar-Faisal JL. In Vitro Evaluation of the Anti-Chikungunya Virus Activity of an Active Fraction Obtained from Euphorbia grandicornis Latex. Viruses 2024; 16:1929. [PMID: 39772236 PMCID: PMC11680167 DOI: 10.3390/v16121929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/14/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Chikungunya virus (CHIKV) is classified as a pathogen with the potential to cause a pandemic. This situation becomes more alarming since no approved drug exists to combat the virus. The present research aims to demonstrate the anti-CHIKV activity of molecules present in the latex of Euphorbia grandicornis. Therefore, a biodirected assay was carried out to find the molecules with anti-CHIKV activity. Extractions with hexane, dichloromethane, and methanol and subsequent purification by column chromatography were carried out to later evaluate cytotoxic activity by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay and antiviral activity by plaque assay. Our findings show that unlike the others, methanolic extract has a low cytotoxic effect and a good anti-CHIKV effect (EC50 = 26.41 µg/mL), which increases when obtaining the purified active fraction (pAFeg1) (EC50 = 0.4835 µg/mL). Time-of-addition suggests that the possible mechanism of action of pAFeg1 could be inhibiting any of the non-structural proteins of CHIKV. In addition, both the cytotoxic and anti-CHIKV activity of pAFeg1 demonstrate selectivity since it killed cancer cells and could not inhibit DENV2.
Collapse
Affiliation(s)
- José Angel Santiago-Cruz
- Laboratorio de Medicina de Conservación de la Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis, Colonia Casco de Santo Tomas, Ciudad de Mexico 11340, Mexico; (A.P.-M.); (A.P.-J.); (J.E.A.-G.)
| | - Araceli Posadas-Mondragón
- Laboratorio de Medicina de Conservación de la Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis, Colonia Casco de Santo Tomas, Ciudad de Mexico 11340, Mexico; (A.P.-M.); (A.P.-J.); (J.E.A.-G.)
| | - Angélica Pérez-Juárez
- Laboratorio de Medicina de Conservación de la Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis, Colonia Casco de Santo Tomas, Ciudad de Mexico 11340, Mexico; (A.P.-M.); (A.P.-J.); (J.E.A.-G.)
| | - Norma Estela Herrera-González
- Laboratorio de Oncología Molecular de la Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis, Colonia Casco de Santo Tomas, Ciudad de Mexico 11340, Mexico;
| | - José Miguel Chin-Chan
- Laboratorio de Epigenética Ambiental y Salud Mental, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Campeche, Ciudad de Campeche 24039, Mexico;
| | - Joab Eli Aguilar-González
- Laboratorio de Medicina de Conservación de la Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis, Colonia Casco de Santo Tomas, Ciudad de Mexico 11340, Mexico; (A.P.-M.); (A.P.-J.); (J.E.A.-G.)
| | - José Leopoldo Aguilar-Faisal
- Laboratorio de Medicina de Conservación de la Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis, Colonia Casco de Santo Tomas, Ciudad de Mexico 11340, Mexico; (A.P.-M.); (A.P.-J.); (J.E.A.-G.)
| |
Collapse
|
5
|
Resck MEB, Câmara DCP, dos Santos FB, dos Santos JPC, Alto BW, Honório NA. Spatial-temporal distribution of chikungunya virus in Brazil: a review on the circulating viral genotypes and Aedes ( Stegomyia) albopictus as a potential vector. Front Public Health 2024; 12:1496021. [PMID: 39722706 PMCID: PMC11668782 DOI: 10.3389/fpubh.2024.1496021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Chikungunya virus (CHIKV) is mainly transmitted by the invasive mosquito Aedes (Stegomyia) aegypti in tropical and subtropical regions worldwide. However, genetic adaptations of the virus to the peri domestic mosquito vector Aedes (Stegomyia) albopictus has resulted in enhanced vector competence and associated epidemics and may contribute to further geographic expansion of CHIKV. However, evidence-based data on the relative role of Ae. albopictus in CHIKV transmission dynamics are scarce, especially in regions where Ae. aegypti is the main vector, such as in Brazil. Here, we review the CHIKV genotypes circulating in Brazil, spatial and temporal distribution of Chikungunya cases in Brazil, and susceptibility to infection and transmission (i.e., vector competence) of Ae. albopictus for CHIKV to better understand its relative contribution to the virus transmission dynamics.
Collapse
Affiliation(s)
| | - Daniel Cardoso Portela Câmara
- Programa de Computação Científica, Fundação Oswaldo Cruz - PROCC, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Flávia Barreto dos Santos
- Laboratório das Interações Vírus-Hospedeiros - LIVH, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, Brazil
| | | | - Barry Wilmer Alto
- Florida Medical Entomology Laboratory-FMEL, University of Florida, Vero Beach, FL, United States
| | - Nildimar Alves Honório
- Laboratório das Interações Vírus-Hospedeiros - LIVH, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, Brazil
- Núcleo Operacional Sentinela de Mosquitos Vetores-Nosmove/Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
6
|
Bullock CB, Wang L, Ware BC, Wagoner N, Ohara RA, Liu TT, Desai P, Peters B, Murphy KM, Handley SA, Morrison TE, Diamond MS. Type I interferon signaling in dendritic cells limits direct antigen presentation and CD8 + T cell responses against an arthritogenic alphavirus. mBio 2024; 15:e0293024. [PMID: 39535221 PMCID: PMC11633147 DOI: 10.1128/mbio.02930-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Ross River virus (RRV) and other alphaviruses cause chronic musculoskeletal syndromes that are associated with viral persistence, which suggests deficits in immune clearance mechanisms, including CD8+ T-cell responses. Here, we used a recombinant RRV-gp33 that expresses the immunodominant CD8+ T-cell epitope of lymphocytic choriomeningitis virus (LCMV) to directly compare responses with a virus, LCMV, that strongly induces antiviral CD8+ T cells. After footpad injection, we detected fewer gp33-specific CD8+ T cells in the draining lymph node (DLN) after RRV-gp33 than LCMV infection, despite similar viral RNA levels in the foot. However, less RRV RNA was detected in the DLN compared to LCMV, with RRV localizing principally to the subcapsular region and LCMV to the paracortical T-cell zones. Single-cell RNA-sequencing analysis of adoptively transferred gp33-specific transgenic CD8+ T cells showed rapid differentiation into effector cells after LCMV but not RRV infection. This defect in RRV-specific CD8+ T effector cell maturation was corrected by local blockade of type I interferon (IFN) signaling, which also resulted in increased RRV infection in the DLN. Studies in Wdfy4-/-, CD11c-Cre B2mfl/fl, or Xcr1-Cre Ifnar1fl/fl mice that respectively lack cross-presenting capacity, MHC-I antigen presentation by dendritic cells (DCs), or type I IFN signaling in the DC1 subset show that RRV-specific CD8+ T-cell responses can be improved by enhanced direct antigen presentation by DCs. Overall, our experiments suggest that antiviral type I IFN signaling in DCs limits direct alphavirus infection and antigen presentation, which likely delays CD8+ T-cell responses.IMPORTANCEChronic arthritis and musculoskeletal disease are common outcomes of infections caused by arthritogenic alphaviruses, including Ross River virus (RRV), due to incomplete virus clearance. Unlike other viral infections that are efficiently cleared by cytotoxic CD8+ T cells, RRV infection is surprisingly unaffected by CD8+ T cells as mice lacking or having these cells show similar viral persistence in joint and lymphoid tissues. To elucidate the basis for this deficient response, we measured the RRV-specific CD8+ T-cell population size and activation state relative to another virus known to elicit a strong T-cell response. Our findings reveal that RRV induces fewer CD8+ T cells due to limited infection of immune cells in the draining lymph node. By increasing RRV susceptibility in antigen-presenting cells, we elicited a robust CD8+ T-cell response. These results highlight antigen availability and virus tropism as possible targets for intervention against RRV immune evasion and persistence.
Collapse
Affiliation(s)
- Christopher B. Bullock
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Leran Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Brian C. Ware
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Ngan Wagoner
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ray A. Ohara
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tian-Tian Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Pritesh Desai
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Bjoern Peters
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Kenneth M. Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Scott A. Handley
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Thomas E. Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Michael S. Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
7
|
Sridhar S, Tonto PB, Lumkong L, Netto EM, Brites C, Wang WK, Herrera BB. RT-RPA as a dual tool for detection and phylogenetic analysis of epidemic arthritogenic alphaviruses. Sci Rep 2024; 14:30134. [PMID: 39627454 PMCID: PMC11615341 DOI: 10.1038/s41598-024-81763-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 11/28/2024] [Indexed: 12/06/2024] Open
Abstract
Chikungunya (CHIKV), o'nyong-nyong (ONNV), and Mayaro (MAYV) viruses are transmitted by mosquitoes and known to cause a debilitating arthritogenic syndrome. These alphaviruses have emerged and re-emerged, leading to outbreaks in tropical and subtropical regions of Asia, South America, and Africa. Despite their prevalence, there persists a critical gap in the availability of sensitive and virus-specific point-of-care (POC) diagnostics. Traditional immunoglobulin-based tests such as enzyme-linked immunosorbent assay (ELISA) often yield cross-reactive results due to the close genetic relationship between these viruses. Molecular diagnostics such as quantitative polymerase chain reaction (qPCR) offer high sensitivity but are limited by the need for specialized laboratory equipment. Recombinase polymerase amplification (RPA), an isothermal amplification method, is a promising alternative to qPCR, providing rapid results with minimal equipment requirements. Here, we report the development and validation of three virus-specific RT-RPA-based rapid tests for CHIKV, ONNV, and MAYV. These tests demonstrated both speed and sensitivity, capable of detecting 10-100 viral copies within 20 min of amplification, without exhibiting cross-reactivity. Furthermore, we evaluated the clinical potential of these tests using serum and tissue samples from CHIKV, ONNV, and MAYV-infected mice, as well as CHIKV-infected human patients. We demonstrate that the RPA amplicons derived from the patient samples can be sequenced, enabling cost-effective molecular epidemiological studies. Our findings highlight the significance of these rapid and specific diagnostics in improving the early detection and management of these arboviral infections, particularly in resource-limited settings.
Collapse
Affiliation(s)
- Sainetra Sridhar
- Rutgers Global Health Institute, Rutgers University, New Brunswick, NJ, USA
- Department of Medicine, Division of Allergy, Immunology, and Infectious Diseases, and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Prince Baffour Tonto
- Rutgers Global Health Institute, Rutgers University, New Brunswick, NJ, USA
- Department of Medicine, Division of Allergy, Immunology, and Infectious Diseases, and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Lily Lumkong
- Rutgers Global Health Institute, Rutgers University, New Brunswick, NJ, USA
- Department of Medicine, Division of Allergy, Immunology, and Infectious Diseases, and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Eduardo Martins Netto
- School of Medicine, LAPI-Laboratório de Pesquisa em Infectologia- Hospital Universitário Professor Edgard Santos, Federal University of Bahia/EBSERH, Salvador, Brazil
| | - Carlos Brites
- School of Medicine, LAPI-Laboratório de Pesquisa em Infectologia- Hospital Universitário Professor Edgard Santos, Federal University of Bahia/EBSERH, Salvador, Brazil
| | - Wei-Kung Wang
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Bobby Brooke Herrera
- Rutgers Global Health Institute, Rutgers University, New Brunswick, NJ, USA.
- Department of Medicine, Division of Allergy, Immunology, and Infectious Diseases, and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
8
|
de Roo AM, Vondeling GT, Boer M, Murray K, Postma MJ. The global health and economic burden of chikungunya from 2011 to 2020: a model-driven analysis on the impact of an emerging vector-borne disease. BMJ Glob Health 2024; 9:e016648. [PMID: 39627007 PMCID: PMC11624783 DOI: 10.1136/bmjgh-2024-016648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/14/2024] [Indexed: 12/09/2024] Open
Abstract
INTRODUCTION Chikungunya is a mosquito-borne arboviral disease posing an emerging global public health threat. Understanding the global burden of chikungunya is critical for designing effective prevention and control strategies. However, current estimates of the economic and health impact of chikungunya remain limited and are potentially underestimated. This study aims to provide a comprehensive overview of the chikungunya burden worldwide. METHODS We analysed the global burden of chikungunya between 2011 and 2020 and calculated disability-adjusted life years (DALYs) and direct and indirect costs using a data-driven simulation model. The main outcomes were the number of cases, the total DALY burden, and the direct and indirect costs of acute and chronic chikungunya between 2011 and 2020. RESULTS Our study revealed a total of 18.7 million chikungunya cases in 110 countries between 2011 and 2020, causing 1.95 million DALYs. Most of this burden was found in the Latin American and Caribbean region. The total economic burden caused by chikungunya over these 10 years was estimated at $2.8 billion in direct costs and $47.1 billion in indirect costs worldwide. Long-term chronic illness was the source of most costs and DALYs. CONCLUSION Chikungunya has a higher disease burden than was previously estimated and costs related to the disease are substantial. Especially in combination with its unpredictable nature, chikungunya could significantly impact local health systems. Insights from this study could inform decision makers on the impact of chikungunya on population health and help them to appropriately allocate resources to protect vulnerable populations from this debilitating disease.
Collapse
Affiliation(s)
- Adrianne Marije de Roo
- Valneva Austria GmbH, Vienna, Austria
- Department of Health Sciences, University of Groningen, Groningen, Netherlands
| | | | - Martijn Boer
- ASC Academics BV, Groningen, Groningen, Netherlands
| | - Kristy Murray
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Maarten Jacobus Postma
- Department of Health Sciences, University of Groningen, Groningen, Groningen, Netherlands
- Department of Economics, Econometrics & Finance, University of Groningen, Groningen, Netherlands
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Badung, Indonesia
- Division of Pharmacology and Therapy, Faculty of Medicine Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
9
|
Yeh-Gorocica A, Torres-Castro M, Carrillo-Chan C, Suarez-Galaz A, Suarez-Galaz M, Moguel-Chin W, Panti-May A, Lugo-Caballero C, Puerta-Guardo H, Chable-Santos J, Manrique-Saide P, Ayora-Talavera G, Selem-Salas C, Frias-Casas M, Rivero-Juarez A. Prevalence of Flavivirus and A lphavirus in bats captured in the state of Yucatan, southeastern Mexico. One Health 2024; 19:100876. [PMID: 39258264 PMCID: PMC11386310 DOI: 10.1016/j.onehlt.2024.100876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024] Open
Abstract
Flavivirus (family Flaviviridae) and Alphavirus (family Togaviridae) are mosquito-borne viruses that poses a significant risk to public health worldwide. Examples of these viruses include Dengue virus (DENV) and Zika virus (ZIKV) in the Flavivirus genus, and Chikungunya virus (CHIKV) in the Alphavirus genus. The potential contribution of bats in the mosquito-to-human transmission cycle of these viral genera in the tropics has not been studied. Here, a total of 144 bats belonging to three families (Emballonuridae, Phyllostomidae, and Molossidae) and six species were captured for one year using mist nets in sites with different landscapes (forest and grassland) in the state of Yucatan, southeastern Mexico. Blood samples and rectal and oral swabs were collected to detect Flavivirus and Alphavirus RNA genomes through RT-PCR. Flavivirus RNA was detected in 53 individuals (36.8%; 95% CI: 29.4%-44.9%), and Alphavirus RNA was detected in 59 individuals (40.1%; 95% CI: 33.2%-49.2%). The sequences obtained were consistent with ZIKV and DENV, into the Flavivirus, and CHIKV into the Alphavirus positive samples. The prevalence of both Flavivirus and Alphavirus was higher during the dry season compared with the rainy season. This high positivity rate, highlighted in both Flavivirus and Alphavirus, suggests a potential contribution of bats in the circulation of these viral genera in sylvatic environments. Seasonal variation in viral genera prevalence, with higher prevalence during dry seasons than rainy seasons, may suggest specific viral activity patterns in response to climatic conditions.
Collapse
Affiliation(s)
- Aaron Yeh-Gorocica
- Laboratorio de Zoonosis y otras Enfermedades Transmitidas por Vector, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autonoma de Yucatan, Merida, Mexico
| | - Marco Torres-Castro
- Laboratorio de Zoonosis y otras Enfermedades Transmitidas por Vector, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autonoma de Yucatan, Merida, Mexico
| | - Claudia Carrillo-Chan
- Laboratorio de Zoonosis y otras Enfermedades Transmitidas por Vector, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autonoma de Yucatan, Merida, Mexico
| | - Alejandro Suarez-Galaz
- Laboratorio de Zoonosis y otras Enfermedades Transmitidas por Vector, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autonoma de Yucatan, Merida, Mexico
| | - Melissa Suarez-Galaz
- Laboratorio de Zoonosis y otras Enfermedades Transmitidas por Vector, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autonoma de Yucatan, Merida, Mexico
| | - Wilson Moguel-Chin
- Laboratorio de Zoonosis y otras Enfermedades Transmitidas por Vector, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autonoma de Yucatan, Merida, Mexico
| | - Alonso Panti-May
- Laboratorio de Zoonosis y otras Enfermedades Transmitidas por Vector, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autonoma de Yucatan, Merida, Mexico
| | - Cesar Lugo-Caballero
- Laboratorio de Enfermedades Emergentes y Reemergentes, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autonoma de Yucatan, Merida, Mexico
| | - Henry Puerta-Guardo
- Laboratorio de Virologia, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autonoma de Yucatan, Merida, Mexico
- Unidad Colaborativa para Bioensayos Entomologicos, Facultad de Medicina Veterinaria y Zootecnia, Campus de Ciencias Biologicas y Agropecuarias, Universidad Autonoma de Yucatan, Merida, Mexico
| | - Juan Chable-Santos
- Departamento de Zoologia, Facultad de Medicina Veterinaria y Zootecnia, Campus de Ciencias Biologicas y Agropecuarias, Universidad Autonoma de Yucatan, Merida, Mexico
| | - Pablo Manrique-Saide
- Departamento de Zoologia, Facultad de Medicina Veterinaria y Zootecnia, Campus de Ciencias Biologicas y Agropecuarias, Universidad Autonoma de Yucatan, Merida, Mexico
- Unidad Colaborativa para Bioensayos Entomologicos, Facultad de Medicina Veterinaria y Zootecnia, Campus de Ciencias Biologicas y Agropecuarias, Universidad Autonoma de Yucatan, Merida, Mexico
| | - Guadalupe Ayora-Talavera
- Laboratorio de Virologia, Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autonoma de Yucatan, Merida, Mexico
| | - Celia Selem-Salas
- Departamento de Zoologia, Facultad de Medicina Veterinaria y Zootecnia, Campus de Ciencias Biologicas y Agropecuarias, Universidad Autonoma de Yucatan, Merida, Mexico
| | - Mario Frias-Casas
- Virologia Clinica y Zoonosis, Instituto Maimonides de Investigacion Biomedica de Cordoba (IMIBIC), Hospital Universitario Reina Sofia de Cordoba, Universidad de Cordoba, Cordoba, Spain
- CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Rivero-Juarez
- Virologia Clinica y Zoonosis, Instituto Maimonides de Investigacion Biomedica de Cordoba (IMIBIC), Hospital Universitario Reina Sofia de Cordoba, Universidad de Cordoba, Cordoba, Spain
- CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
10
|
Ning X, Xia B, Wang J, Gao R, Ren H. Host-adaptive mutations in Chikungunya virus genome. Virulence 2024; 15:2401985. [PMID: 39263937 PMCID: PMC11404619 DOI: 10.1080/21505594.2024.2401985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/08/2024] [Accepted: 08/31/2024] [Indexed: 09/13/2024] Open
Abstract
Chikungunya virus (CHIKV) is the causative agent of chikungunya fever (CHIKF), and its primary vectors are the mosquitoes Aedes aegypti and Aedes albopictus. CHIKV was initially endemic to Africa but has spread globally in recent years and affected millions of people. According to a risk assessment by the World Health Organization, CHIKV has the potential seriously impact public health. A growing body of research suggests that mutations in the CHIKV gene that enhance viral fitness in the host are contributing to the expansion of the global CHIKF epidemic. In this article, we review the host-adapted gene mutations in CHIKV under natural evolution and laboratory transmission conditions, which can help improve our understanding of the adaptive evolution of CHIKV and provide a basis for monitoring and early warning of future CHIKV outbreaks.
Collapse
Affiliation(s)
- Xinhang Ning
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, People’s Republic of China
| | - Binghui Xia
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, People’s Republic of China
| | - Jiaqi Wang
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, People’s Republic of China
| | - Rong Gao
- Department of Respiratory Medicine, The People’s Liberation Army Joint Logistic Support Force 943 Hospital, Wuwei, Gansu, People’s Republic of China
| | - Hao Ren
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
11
|
Freppel W, Silva LA, Stapleford KA, Herrero LJ. Pathogenicity and virulence of chikungunya virus. Virulence 2024; 15:2396484. [PMID: 39193780 PMCID: PMC11370967 DOI: 10.1080/21505594.2024.2396484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted, RNA virus that causes an often-severe musculoskeletal illness characterized by fever, joint pain, and a range of debilitating symptoms. The virus has re-emerged as a global health threat in recent decades, spreading from its origin in Africa across Asia and the Americas, leading to widespread outbreaks impacting millions of people. Despite more than 50 years of research into the pathogenesis of CHIKV, there is still no curative treatment available. Current management of CHIKV infections primarily involves providing supportive care to alleviate symptoms and improve the patient's quality of life. Given the ongoing threat of CHIKV, there is an urgent need to better understand its pathogenesis. This understanding is crucial for deciphering the mechanisms underlying the disease and for developing effective strategies for both prevention and management. This review aims to provide a comprehensive overview of CHIKV and its pathogenesis, shedding light on the complex interactions of viral genetics, host factors, immune responses, and vector-related factors. By exploring these intricate connections, the review seeks to contribute to the knowledge base surrounding CHIKV, offering insights that may ultimately lead to more effective prevention and management strategies for this re-emerging global health threat.
Collapse
Affiliation(s)
- Wesley Freppel
- Institute for Biomedicine and Glycomics, Gold Coast Campus, Griffith University, Southport, Australia
| | - Laurie A. Silva
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kenneth A. Stapleford
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Lara J. Herrero
- Institute for Biomedicine and Glycomics, Gold Coast Campus, Griffith University, Southport, Australia
| |
Collapse
|
12
|
Allen SW, Ribeiro Dos Santos G, Paul KK, Paul R, Rahman MZ, Alam MS, Rahman M, Al-Amin HM, Vanhomwegen J, Weaver SC, Smull T, Lee KH, Gurley ES, Salje H. Results of a Nationally Representative Seroprevalence Survey of Chikungunya Virus in Bangladesh. J Infect Dis 2024; 230:e1031-e1038. [PMID: 38942731 PMCID: PMC11565896 DOI: 10.1093/infdis/jiae335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/12/2024] [Accepted: 06/26/2024] [Indexed: 06/30/2024] Open
Abstract
There is an increasing global burden from chikungunya virus (CHIKV). Bangladesh reported a major epidemic in 2017, but it was unclear whether there had been prior widespread transmission. We conducted a nationally representative seroprevalence survey in 70 randomly selected communities immediately before the epidemic. We found that 69 of 2938 sampled individuals (2.4%) were seropositive to CHIKV. Seropositivity to dengue virus (adjusted odds ratio, 3.13 [95% confidence interval, 1.86-5.27]), male sex (0.59 [.36-.99]), and community presence of Aedes aegypti mosquitoes (1.80 [1.05-3.0]7) were significantly associated with CHIKV seropositivity. Using a spatial prediction model, we estimated that across the country, 4.99 (95% confidence interval, 4.89-5.08) million people had been previously infected. These findings highlight high population susceptibility before the major outbreak and that previous outbreaks must have been spatially isolated.
Collapse
Affiliation(s)
- Sam W Allen
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | | | - Kishor K Paul
- Kirby Institute, University of New South Wales, Sydney, Australia
- School of Population Health, University of New South Wales, Sydney, New South Wales, Australia
- One Health Laboratory, icddr,b, Dhaka, Bangladesh
| | - Repon Paul
- One Health Laboratory, icddr,b, Dhaka, Bangladesh
- Centre for Big Data Research in Health, University of New South Wales, Sydney, New South Wales, Australia
| | | | | | | | - Hasan Mohammad Al-Amin
- One Health Laboratory, icddr,b, Dhaka, Bangladesh
- QIMR Berghofer Medical Research Institute, The University of Queensland, Herston, Australia
- School of the Environment, The University of Queensland, Herston, Queensland, Australia
| | - Jessica Vanhomwegen
- Institut Pasteur, Université Paris Cité, Unité Environnement et Risques Infectieux, Cellule d'Intervention Biologique d'Urgence (CIBU), 75015 Paris, France
| | - Scott C Weaver
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Taylor Smull
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Maryland, Baltimore, USA
| | - Kyu Han Lee
- Emory Global Health Institute, Emory University, Atlanta, Georgia, USA
| | - Emily S Gurley
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Maryland, Baltimore, USA
| | - Henrik Salje
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
13
|
Tariq A, Khan A, Mutuku F, Ndenga B, Bisanzio D, Grossi-Soyster EN, Jembe Z, Maina P, Chebii P, Ronga C, Okuta V, LaBeaud AD. Understanding the factors contributing to dengue virus and chikungunya virus seropositivity and seroconversion among children in Kenya. PLoS Negl Trop Dis 2024; 18:e0012616. [PMID: 39565798 PMCID: PMC11578454 DOI: 10.1371/journal.pntd.0012616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 10/08/2024] [Indexed: 11/22/2024] Open
Abstract
Dengue virus (DENV) and chikungunya virus (CHIKV) are causes of endemic febrile disease among Kenyan children. The exposure risk to these infections is highly multifactorial and linked to environmental factors and human behavior. We investigated relationships between household, socio-economic, demographic, and behavioral risk factors for DENV and CHIKV seropositivity and seroconversion in four settlements in Kenya. We prospectively followed a pediatric cohort of 3,445 children between 2014-2018. We utilized the Kaplan-Meier curves to describe the temporal patterns of seroconversion among tested participants. We employed logistic regression built using generalized linear mixed models, to identify potential exposure risk factors for DENV and CHIKV seroconversion and seropositivity. Overall, 5.2% children were seropositive for DENV, of which 59% seroconverted during the study period. The seroprevalence for CHIKV was 9.2%, of which 54% seroconverted. The fraction of seroconversions per year in the study cohort was <2% for both viruses. Multivariable analysis indicated that older age and the presence of water containers ((OR: 1.15 [95% CI: 1.10, 1.21]), (OR: 1.50 [95% CI: 1.07, 2.10])) increased the odds of DENV seropositivity, whereas higher wealth (OR: 0.83 [95% CI: 0.73, 0.96]) decreased the odds of DENV seropositivity. Multivariable analysis for CHIKV seropositivity showed older age and the presence of trash in the housing compound to be associated with increased odds of CHIKV seropositivity ((OR: 1.11[95% CI: 1.07, 1.15]), (OR: 1.34 [95% CI: 1.04, 1.73])), while higher wealth decreased the odds of CHIKV seropositivity (OR: 0.74[95% CI: 0.66, 0.83]). A higher wealth index (OR: 0.82 [95% CI: 0.69, 0.97]) decreased the odds of DENV seroconversion, whereas a higher age (OR: 1.08 [95% CI: 1.02, 1.15]) and the presence of water containers in the household (OR: 1.91[95% CI: 1.24, 2.95]) were significantly associated with increased odds of DENV seroconversion. Higher wealth was associated with decreased odds for CHIKV seroconversion (OR: 0.75 [95% CI: 0.66, 0.89]), whereas presence of water containers in the house (OR: 1.57 [95% CI: 1.11, 2.21]) was a risk factor for CHIKV seroconversion. Our study links ongoing CHIKV and DENV exposure to decreased wealth and clean water access, underscoring the need to combat inequity and poverty and further enhance ongoing surveillance for arboviruses in Kenya to decrease disease transmission. The study emphasizes the co-circulation of DENV and CHIKV and calls for strengthening the targeted control strategies of mosquito borne diseases in Kenya including vector control, environmental management, public education, community engagement and personal protection.
Collapse
Affiliation(s)
- Amna Tariq
- Department of Pediatrics, Division of Infectious Diseases, Stanford University, Palo Alto, California, United States of America
| | - Aslam Khan
- Department of Pediatrics, Division of Infectious Diseases, Stanford University, Palo Alto, California, United States of America
| | - Francis Mutuku
- Department of Environment and Health Sciences, Technical University of Mombasa, Mombasa, Kenya
| | - Bryson Ndenga
- Centre for Global Health Research, Kenya, Medical Research Institute, Kisumu, Kenya
| | - Donal Bisanzio
- RTI International, Washington, D.C, United States of America
| | - Elysse N. Grossi-Soyster
- Department of Pediatrics, Division of Infectious Diseases, Stanford University, Palo Alto, California, United States of America
| | - Zainab Jembe
- Vector borne Disease control Unit, Msambweni County Referral Hospital, Msambweni, Kenya
| | - Priscilla Maina
- Vector borne Disease control Unit, Msambweni County Referral Hospital, Msambweni, Kenya
| | - Philip Chebii
- Vector borne Disease control Unit, Msambweni County Referral Hospital, Msambweni, Kenya
| | - Charles Ronga
- Centre for Global Health Research, Kenya, Medical Research Institute, Kisumu, Kenya
| | - Victoria Okuta
- Centre for Global Health Research, Kenya, Medical Research Institute, Kisumu, Kenya
| | - Angelle Desiree LaBeaud
- Department of Pediatrics, Division of Infectious Diseases, Stanford University, Palo Alto, California, United States of America
| |
Collapse
|
14
|
Ali Mude AS, Nageye YA, Bello KE. Current Epidemiological Status of Chikungunya Virus Infection in East Africa: A Systematic Review and Meta-Analysis. J Trop Med 2024; 2024:7357911. [PMID: 39492843 PMCID: PMC11530290 DOI: 10.1155/2024/7357911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
Background: The incidence of Chikungunya in tropical Africa is still of major epidemiological significance. This study aims to determine the prevalence of chikungunya in East Africa through a systematic review and meta-analysis of published studies. Methods: We conducted a comprehensive search across six electronic databases-Web of Science, PubMed, ScienceDirect, Scopus, and Google Scholar-using specific keywords to address the worldwide impact of chikungunya following the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guidelines. A meta-analysis was performed on our eligible studies using the random effect model. Results: Our search returned 40 eligible articles involving 4122 Chikungunya cases in 13 East African nations. These studies, conducted between 2014 and 2024 across 13 East African nations, provided diverse data on chikungunya prevalence. The overall pooled prevalence of chikungunya in East Africa was 20.6% (95% CI: 18.8%-22.5% and I 2 = 99.62%). Subgroup analyses revealed variations in prevalence across different countries, study designs, detection methods, and publication years. Notably, Rwanda and Djibouti exhibited high prevalence rates of 63.0% and 50.4%, respectively, while Kenya and Somalia reported a moderate prevalence of 12.2%. The detection methods also influenced prevalence rates, with RT-PCR studies indicating a higher prevalence (28.3%) compared to ELISA (19.3%). Conclusion: The study highlights the significant burden of chikungunya in East Africa, and the findings underscore the need for targeted public health interventions and improved surveillance to manage and control chikungunya outbreaks in the region.
Collapse
Affiliation(s)
- Abdirasak Sharif Ali Mude
- Department of Microbiology and Laboratory Science, Faculty of Medicine and Health Sciences, SIMAD University, Mogadishu 252, Somalia
| | - Yahye Ahmed Nageye
- Department of Microbiology and Laboratory Science, Faculty of Medicine and Health Sciences, SIMAD University, Mogadishu 252, Somalia
| | - Kizito Eneye Bello
- Department of Microbiology, Faculty of Natural Science, Kogi State (Prince Abubakar Audu) University, Anyigba PMB 1008, Kogi State, Nigeria
| |
Collapse
|
15
|
Martins DOS, Ruiz UEA, Santos IA, Oliveira IS, Guevara-Vega M, de Paiva REF, Abbehausen C, Sabino-Silva R, Corbi PP, Jardim ACG. Exploring the antiviral activities of the FDA-approved drug sulfadoxine and its derivatives against Chikungunya virus. Pharmacol Rep 2024; 76:1147-1159. [PMID: 39150661 DOI: 10.1007/s43440-024-00635-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Currently, there is no antiviral licensed to treat chikungunya fever, a disease caused by the infection with Alphavirus chikungunya (CHIKV). Treatment is based on analgesic and anti-inflammatory drugs to relieve symptoms. Our study aimed to evaluate the antiviral activity of sulfadoxine (SFX), an FDA-approved drug, and its derivatives complexed with silver(I) (AgSFX), salicylaldehyde Schiff base (SFX-SL), and with both Ag and SL (AgSFX-SL) against CHIKV. METHODS The anti-CHIKV activity of SFX and its derivatives was investigated using BHK-21 cells infected with CHIKV-nanoluc, a marker virus-carrying nanoluciferase reporter. Dose-response and time of drug-addition assays were performed in order to assess the antiviral effects of the compounds, as well as in silico data and ATR-FTIR analysis for insights on their mechanisms of action. RESULTS The SFX inhibited 34% of CHIKV replication, while AgSFX, SFX-SL, and AgSFX-SL enhanced anti-CHIKV activity to 84%, 89%, and 95%, respectively. AgSFX, SFX-SL, and AgSFX-SL significantly decreased viral entry and post-entry to host cells, and the latter also protected cells against infection. Additionally, molecular docking calculations and ATR-FTIR analysis demonstrated interactions of SFX-SL, AgSFX, and AgSFX-SL with CHIKV. CONCLUSIONS Collectively, our findings suggest that the addition of metal ions and/or Schiff base to SFX improved its antiviral activity against CHIKV.
Collapse
Affiliation(s)
- Daniel Oliveira Silva Martins
- Institute of Biomedical Science - ICBIM, Federal University of Uberlândia - UFU, Avenida Amazonas, 4C- Room 216, Umuarama, Uberlândia, MG, CEP: 38405-302, Brazil
- Institute of Bioscience, Language and Exact Sciences - IBILCE, São Paulo State University - UNESP, São José do Rio Preto, SP, Brazil
| | - Uriel Enrique Aquino Ruiz
- Institute of Biomedical Science - ICBIM, Federal University of Uberlândia - UFU, Avenida Amazonas, 4C- Room 216, Umuarama, Uberlândia, MG, CEP: 38405-302, Brazil
| | - Igor Andrade Santos
- Institute of Biomedical Science - ICBIM, Federal University of Uberlândia - UFU, Avenida Amazonas, 4C- Room 216, Umuarama, Uberlândia, MG, CEP: 38405-302, Brazil
| | | | - Marco Guevara-Vega
- Institute of Biomedical Science - ICBIM, Federal University of Uberlândia - UFU, Avenida Amazonas, 4C- Room 216, Umuarama, Uberlândia, MG, CEP: 38405-302, Brazil
| | | | - Camilla Abbehausen
- Institute of Chemistry, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Robinson Sabino-Silva
- Institute of Biomedical Science - ICBIM, Federal University of Uberlândia - UFU, Avenida Amazonas, 4C- Room 216, Umuarama, Uberlândia, MG, CEP: 38405-302, Brazil
| | - Pedro Paulo Corbi
- Institute of Chemistry, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Ana Carolina Gomes Jardim
- Institute of Biomedical Science - ICBIM, Federal University of Uberlândia - UFU, Avenida Amazonas, 4C- Room 216, Umuarama, Uberlândia, MG, CEP: 38405-302, Brazil.
- Institute of Bioscience, Language and Exact Sciences - IBILCE, São Paulo State University - UNESP, São José do Rio Preto, SP, Brazil.
| |
Collapse
|
16
|
Graham VA, Easterbrook L, Rayner E, Findlay-Wilson S, Flett L, Kennedy E, Fotheringham S, Kempster S, Almond N, Dowall S. Comparison of Chikungunya Virus-Induced Disease Progression and Pathogenesis in Type-I Interferon Receptor-Deficient Mice (A129) and Two Wild-Type (129Sv/Ev and C57BL/6) Mouse Strains. Viruses 2024; 16:1534. [PMID: 39459867 PMCID: PMC11512278 DOI: 10.3390/v16101534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus causing a debilitating febrile illness with rheumatic disease symptoms of arthralgia and arthritis. Since its spread outside of Africa in 2005, it continues to cause outbreaks and disseminates into new territories. Intervention strategies are urgently required, including vaccination and antiviral approaches. To test efficacy, the use of small animal models is required. Two mouse strains, A129, with a deficiency in their type-I interferon (IFN) receptor, and C57BL/6 are widely used. A direct comparison of these strains alongside the wild-type parental strain of the A129 mice, 129Sv/Ev, was undertaken to assess clinical disease progression, viral loads in key tissues, histological changes and levels of sera biomarkers. Our results confirm the severe disease course in A129 mice which was not observed in the parental 129Sv/Ev strain. Of the two wild-type strains, viral loads were higher in 129Sv/Ev mice compared to C57BL/6 counterparts. Our results have established these models and parameters for the future testing of vaccines and antiviral approaches.
Collapse
Affiliation(s)
- Victoria A. Graham
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, Wiltshire, UK; (V.A.G.); (L.E.); (E.R.); (S.F.-W.); (L.F.); (E.K.); (S.F.)
| | - Linda Easterbrook
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, Wiltshire, UK; (V.A.G.); (L.E.); (E.R.); (S.F.-W.); (L.F.); (E.K.); (S.F.)
| | - Emma Rayner
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, Wiltshire, UK; (V.A.G.); (L.E.); (E.R.); (S.F.-W.); (L.F.); (E.K.); (S.F.)
| | - Stephen Findlay-Wilson
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, Wiltshire, UK; (V.A.G.); (L.E.); (E.R.); (S.F.-W.); (L.F.); (E.K.); (S.F.)
| | - Lucy Flett
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, Wiltshire, UK; (V.A.G.); (L.E.); (E.R.); (S.F.-W.); (L.F.); (E.K.); (S.F.)
| | - Emma Kennedy
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, Wiltshire, UK; (V.A.G.); (L.E.); (E.R.); (S.F.-W.); (L.F.); (E.K.); (S.F.)
| | - Susan Fotheringham
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, Wiltshire, UK; (V.A.G.); (L.E.); (E.R.); (S.F.-W.); (L.F.); (E.K.); (S.F.)
| | - Sarah Kempster
- Medicines and Healthcare Products Regulatory Agency (MHRA), Blanche Ln, South Mimms, Potters Bar EN6 3QG, Hertfordshire, UK; (S.K.); (N.A.)
| | - Neil Almond
- Medicines and Healthcare Products Regulatory Agency (MHRA), Blanche Ln, South Mimms, Potters Bar EN6 3QG, Hertfordshire, UK; (S.K.); (N.A.)
| | - Stuart Dowall
- UK Health Security Agency (UKHSA), Porton Down, Salisbury SP4 0JG, Wiltshire, UK; (V.A.G.); (L.E.); (E.R.); (S.F.-W.); (L.F.); (E.K.); (S.F.)
| |
Collapse
|
17
|
Salles TS, Martins-Duarte ES, de Meneses MDF, Moreira MF, Ferreira DF, Azevedo RC, De Souza W, Caldas LA. Temperature Interference on ZIKV and CHIKV Cycles in Mosquitoes and Mammalian Cells. Pathogens 2024; 13:814. [PMID: 39339005 PMCID: PMC11435172 DOI: 10.3390/pathogens13090814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Temperature is a determining factor for the viral cycle. In this study, we investigate the effect of different temperatures on the cycles of two important arboviruses-Zika (ZIKV) and Chikungunya (CHIKV)-in Vero (mammalian) and C6/36 (mosquito) cells. We compare genome quantification to infectivity at 28 °C and 37 °C in both cell types. Virus-cell interaction was also examined by transmission electron microscopy, allowing the observation of phenomena such as virus-surfing and giant forms for CHIKV, as well as the the scarcity of ZIKV in C6/36 cells compared to its cycle in mammalian cells.
Collapse
Affiliation(s)
- Tiago Souza Salles
- Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro 21941-909, Brazil; (T.S.S.); (M.F.M.)
- Fiocruz Biodiversity and Health Biobank, Oswaldo Cruz Foundation, Rio de Janeiro 21040-361, Brazil
| | | | - Marcelo Damião Ferreira de Meneses
- Department of Virology, Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (M.D.F.d.M.); (R.C.A.)
| | - Monica Ferreira Moreira
- Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro 21941-909, Brazil; (T.S.S.); (M.F.M.)
| | | | - Renata Campos Azevedo
- Department of Virology, Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (M.D.F.d.M.); (R.C.A.)
| | - Wanderley De Souza
- Precision Medicine Research Center, Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-904, Brazil;
- National Centre for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Lucio Ayres Caldas
- Precision Medicine Research Center, Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-904, Brazil;
- Multidisciplinary Research Centre (Numpex-Bio), Federal University of Rio de Janeiro, Rio de Janeiro 25265-970, Brazil
| |
Collapse
|
18
|
Zheng X, He Y, Xia B, Tang W, Zhang C, Wang D, Tang H, Zhao P, Peng H, Liu Y. Etravirine Prevents West Nile Virus and Chikungunya Virus Infection Both In Vitro and In Vivo by Inhibiting Viral Replication. Pharmaceutics 2024; 16:1111. [PMID: 39339151 PMCID: PMC11435157 DOI: 10.3390/pharmaceutics16091111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
Diseases transmitted by arthropod-borne viruses such as West Nile virus (WNV) and chikungunya virus (CHIKV) pose threat to global public health. Unfortunately, to date, there is no available approved drug for severe symptoms caused by both viruses. It has been reported that reverse transcriptase inhibitors can effectively inhibit RNA polymerase activity of RNA viruses. We screened the anti-WNV activity of the FDA-approved reverse transcriptase inhibitor library and found that 4 out of 27 compounds showed significant antiviral activity. Among the candidates, etravirine markedly inhibited WNV infection in both Huh 7 and SH-SY5Y cells. Further assays revealed that etravirine inhibited the infection of multiple arboviruses, including yellow fever virus (YFV), tick-borne encephalitis virus (TBEV), and CHIKV. A deeper study at the phase of action showed that the drug works primarily during the viral replication process. This was supported by the strong interaction potential between etravirine and the RNA-dependent RNA polymerase (RdRp) of WNV and alphaviruses, as evaluated using molecular docking. In vivo, etravirine significantly rescued mice from WNV infection-induced weight loss, severe neurological symptoms, and death, as well as reduced the viral load and inflammatory cytokines in target tissues. Etravirine showed antiviral effects in both arthrophlogosis and lethal mouse models of CHIKV infection. This study revealed that etravirine is an effective anti-WNV and CHIKV arbovirus agent both in vitro and in vivo due to the inhibition of viral replication, providing promising candidates for clinical application.
Collapse
Affiliation(s)
- Xu Zheng
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Yanhua He
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Binghui Xia
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Wanda Tang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Congcong Zhang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Dawei Wang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Hailin Tang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Ping Zhao
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Haoran Peng
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Yangang Liu
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (X.Z.); (Y.H.); (B.X.); (W.T.); (C.Z.); (D.W.); (H.T.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
19
|
Liu Y, Xu M, Xia B, Qiao Z, He Y, Liu Y, Pan Z, Zhang C, Peng H, Liang X, Zhao P, Tang H, Zheng X. Nifuroxazide Prevents Chikungunya Virus Infection Both In Vitro and In Vivo via Suppressing Viral Replication. Viruses 2024; 16:1322. [PMID: 39205296 PMCID: PMC11360488 DOI: 10.3390/v16081322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
Chikungunya virus (CHIKV) is a reemerging arbovirus causing disease on a global scale, and the potential for its epidemics remains high. CHIKV has caused millions of cases and heavy economic burdens around the world, while there are no available approved antiviral therapies to date. In this study, nifuroxazide, an FDA-approved antibiotic for acute diarrhea or colitis, was found to significantly inhibit a variety of arboviruses, although its antiviral activity varied among different target cell types. Nifuroxazide exhibited relatively high inhibitory efficiency in yellow fever virus (YFV) infection of the hepatoma cell line Huh7, tick-borne encephalitis virus (TBEV) and west nile virus (WNV) infection of the vascular endothelial cell line HUVEC, and CHIKV infection of both Huh7 cells and HUVECs, while it barely affected the viral invasion of neurons. Further systematic studies on the action stage of nifuroxazide showed that nifuroxazide mainly inhibited in the viral replication stage. In vivo, nifuroxazide significantly reduced the viral load in muscles and protected mice from CHIKV-induced footpad swelling, an inflammation injury within the arthrosis of infected mice. These results suggest that nifuroxazide has a potential clinical application as an antiviral drug, such as in the treatment of CHIKV infection.
Collapse
Affiliation(s)
- Yangang Liu
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Mingxiao Xu
- Department of Infection Diseases, First Affiliated Hospital of Navy Military Medical University, Shanghai 200433, China; (M.X.); (X.L.)
| | - Binghui Xia
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Zhuoyue Qiao
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming 650500, China;
| | - Yanhua He
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Yan Liu
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Zhendong Pan
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Congcong Zhang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Haoran Peng
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Xuesong Liang
- Department of Infection Diseases, First Affiliated Hospital of Navy Military Medical University, Shanghai 200433, China; (M.X.); (X.L.)
| | - Ping Zhao
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Hailin Tang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| | - Xu Zheng
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; (Y.L.); (B.X.); (Y.H.); (Y.L.); (Z.P.); (C.Z.); (H.P.); (P.Z.)
- Key Laboratory of Biological Defense, Ministry of Education, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
20
|
Sridhar S, Tonto PB, Lumkong L, Netto EM, Brites C, Wang WK, Herrera BB. Development of RT-RPA-based point-of-care tests for epidemic arthritogenic alphaviruses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594209. [PMID: 38826256 PMCID: PMC11142058 DOI: 10.1101/2024.05.14.594209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Chikungunya (CHIKV), o'nyong-nyong (ONNV), and Mayaro (MAYV) viruses are transmitted by mosquitoes and known to cause a debilitating arthritogenic syndrome. These alphaviruses have emerged and re-emerged, leading to outbreaks in tropical and subtropical regions of Asia, South America, and Africa. Despite their prevalence, there persists a critical gap in the availability of sensitive and virus-specific point-of-care (POC) diagnostics. Traditional immunoglobulin-based tests such as enzyme-linked immunosorbent assay (ELISAs) often yield cross-reactive results due to the close genetic relationship between these viruses. Molecular diagnostics such as quantitative polymerase chain reaction (qPCR) offer high sensitivity but are limited by the need for specialized laboratory equipment. Recombinase polymerase amplification (RPA), an isothermal amplification method, is a promising alternative to qPCR, providing rapid results with minimal equipment requirements. Here, we report the development and validation of three virus-specific RPA-based POC tests for CHIKV, ONNV, and MAYV. These tests demonstrated both speed and sensitivity, capable of detecting 10 viral copies within 20 minutes of amplification, without exhibiting cross-reactivity. Furthermore, we evaluated the clinical potential of these tests using serum and tissue samples from CHIKV, ONNV, and MAYV-infected mice, as well as CHIKV-infected human patients. We demonstrate that the RPA amplicons derived from the patient samples can be sequenced, enabling cost-effective molecular epidemiological studies. Our findings highlight the significance of these rapid and specific POC diagnostics in improving the early detection and management of these arboviral infections.
Collapse
|
21
|
Tretyakova I, Joh J, Gearon M, Kraenzle J, Goedeker S, Pignataro A, Alejandro B, Lukashevich IS, Chung D, Pushko P. Live-attenuated CHIKV vaccine with rearranged genome replicates in vitro and induces immune response in mice. PLoS Negl Trop Dis 2024; 18:e0012120. [PMID: 38648230 PMCID: PMC11075892 DOI: 10.1371/journal.pntd.0012120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 05/07/2024] [Accepted: 03/29/2024] [Indexed: 04/25/2024] Open
Abstract
Chikungunya fever virus (CHIKV) is a mosquito-borne alphavirus that causes wide-spread human infections and epidemics in Asia, Africa and recently, in the Americas. CHIKV is considered a priority pathogen by CEPI and WHO. Despite recent approval of a live-attenuated CHIKV vaccine, development of additional vaccines is warranted due to the worldwide outbreaks of CHIKV. Previously, we developed immunization DNA (iDNA) plasmid capable of launching live-attenuated CHIKV vaccine in vivo. Here we report the use of CHIKV iDNA plasmid to prepare a novel, live-attenuated CHIKV vaccine V5040 with rearranged RNA genome. In V5040, genomic RNA was rearranged to encode capsid gene downstream from the glycoprotein genes. Attenuated mutations derived from experimental CHIKV 181/25 vaccine were also engineered into E2 gene of V5040. The DNA copy of rearranged CHIKV genomic RNA with attenuated mutations was cloned into iDNA plasmid pMG5040 downstream from the CMV promoter. After transfection in vitro, pMG5040 launched replication of V5040 virus with rearranged genome and attenuating E2 mutations. Furthermore, V5040 virus was evaluated in experimental murine models for general safety and immunogenicity. Vaccination with V5040 virus subcutaneously resulted in elicitation of CHIKV-specific, virus-neutralizing antibodies. The results warrant further evaluation of V5040 virus with rearranged genome as a novel live-attenuated vaccine for CHIKV.
Collapse
Affiliation(s)
| | - Joongho Joh
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Mary Gearon
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Jennifer Kraenzle
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Sidney Goedeker
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Ava Pignataro
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Brian Alejandro
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Igor S. Lukashevich
- Department of Pharmacology and Toxicology, School of Medicine, and the Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, Kentucky, United States of America
| | - Donghoon Chung
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Peter Pushko
- Medigen, Inc., Frederick, Maryland, United States of America
| |
Collapse
|
22
|
Doran C, Duits AJ, Gerstenbluth I, Tami A, Bailey A. Adaptive coping strategies among individuals living with long-term chikungunya disease: a qualitative study in Curaçao. BMJ Open 2024; 14:e076352. [PMID: 38326245 PMCID: PMC10860096 DOI: 10.1136/bmjopen-2023-076352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 01/19/2024] [Indexed: 02/09/2024] Open
Abstract
OBJECTIVE Long-term chikungunya disease is characterised by persistent rheumatic symptoms following chikungunya virus infection. As there is no specific treatment available, affected individuals need strategies to adapt. However, research on these is scarce. This study aimed to explore which adaptive coping strategies are employed to manage persistent rheumatic symptoms in daily life. SETTING The study was conducted in Curaçao. DESIGN AND PARTICIPANTS An explorative qualitative study was conducted between September and October 2020, among a purposive sample of adults, 19 women and 4 men affected by long-term chikungunya disease. In-depth interviews were semi-structured and transcribed verbatim. The data were analysed using inductive thematic analysis. RESULTS The disease duration for all participants ranged between 68 and 74 months (6 years). In narrating their experiences of coping with long-term chikungunya disease, four themes were identified: (1) learning to live with the disease; (2) resilience for dealing with pain; (3) maintaining positive self-image and attitude; and (4) coping through spirituality. CONCLUSION To live with long-term chikungunya disease with dignity in spite of physical pain and discomfort, participants tried to retain a sense of control of oneself and one's lives, to not let the disease take over, focusing on the positive in their lives, and finding strength and remain hopeful. Interventions such as cognitive-behavioural therapy and mindfulness exercises may be effective in strengthening or regain affected individual's sense of competence and control by fostering adaptive coping skills and resilience. Subsequently, these interventions may improve health-related quality of life when rheumatic symptoms persist following chikungunya virus infection.
Collapse
Affiliation(s)
- Churnalisa Doran
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
| | - Ashley J Duits
- Department of Immunology, Curaçao Biomedical and Health Research Institute, Willemstad, Curaçao
| | - Izzy Gerstenbluth
- Department of Epidemiology, Curaçao Biomedical and Health Research Institute, Willemstad, Curaçao
| | - Adriana Tami
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, Groningen, The Netherlands
| | - Ajay Bailey
- Department of Human Geography and Spatial Planning, University of Utrecht, Utrecht, The Netherlands
| |
Collapse
|
23
|
Ahmed S, Sultana S, Kundu S, Alam SS, Hossan T, Islam MA. Global Prevalence of Zika and Chikungunya Coinfection: A Systematic Review and Meta-Analysis. Diseases 2024; 12:31. [PMID: 38391778 PMCID: PMC10888207 DOI: 10.3390/diseases12020031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
Zika virus (ZIKV) and chikungunya virus (CHIKV) are arthropod-borne viruses with significant pathogenicity, posing a substantial health and economic burden on a global scale. Moreover, ZIKV-CHIKV coinfection imposes additional therapeutic challenges as there is no specific treatment for ZIKV or CHIKV infection. While a growing number of studies have documented the ZIKV-CHIKV coinfection, there is currently a lack of conclusive reports on this coinfection. Therefore, we performed a systematic review and meta-analysis to determine the true statistics of ZIKV-CHIKV coinfection in the global human population. Relevant studies were searched for in PubMed, Scopus, and Google Scholar without limitation in terms of language or publication date. A total of 33 studies containing 41,460 participants were included in this meta-analysis. The study protocol was registered with PROSPERO under the registration number CRD42020176409. The pooled prevalence and confidence intervals of ZIKV-CHIKV coinfection were computed using a random-effects model. The study estimated a combined global prevalence rate of 1.0% [95% CI: 0.7-1.2] for the occurrence of ZIKV-CHIKV coinfection. The region of North America (Mexico, Haiti, and Nicaragua) and the country of Haiti demonstrated maximum prevalence rates of 2.8% [95% CI: 1.5-4.1] and 3.5% [95% CI: 0.2-6.8], respectively. Moreover, the prevalence of coinfection was found to be higher in the paediatric group (2.1% [95% CI: 0.0-4.2]) in comparison with the adult group (0.7% [95% CI: 0.2-1.1]). These findings suggest that the occurrence of ZIKV-CHIKV coinfection varies geographically and by age group. The results of this meta-analysis will guide future investigations seeking to understand the underlying reasons for these variations and the causes of coinfection and to develop targeted prevention and control strategies.
Collapse
Affiliation(s)
- Saleh Ahmed
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Shabiha Sultana
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Shoumik Kundu
- Department of Chemistry and Biochemistry, Texas Tech University, 2500 Broadway St., Lubbock, TX 79409, USA
| | - Sayeda Sadia Alam
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Tareq Hossan
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Md Asiful Islam
- WHO Collaborating Centre for Global Women's Health, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
24
|
Battisti V, Moesslacher J, Abdelnabi R, Leyssen P, Rosales Rosas AL, Langendries L, Aufy M, Studenik C, Kratz JM, Rollinger JM, Puerstinger G, Neyts J, Delang L, Urban E, Langer T. Design, synthesis, and lead optimization of piperazinyl-pyrimidine analogues as potent small molecules targeting the viral capping machinery of Chikungunya virus. Eur J Med Chem 2024; 264:116010. [PMID: 38104375 DOI: 10.1016/j.ejmech.2023.116010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/10/2023] [Accepted: 11/25/2023] [Indexed: 12/19/2023]
Abstract
The worldwide re-emerge of the Chikungunya virus (CHIKV), the high morbidity associated with it, and the lack of an available vaccine or antiviral treatment make the development of a potent CHIKV-inhibitor highly desirable. Therefore, an extensive lead optimization was performed based on the previously reported CHVB compound 1b and the reported synthesis route was optimized - improving the overall yield in remarkably shorter synthesis and work-up time. Hundred analogues were designed, synthesized, and investigated for their antiviral activity, physiochemistry, and toxicological profile. An extensive structure-activity relationship study (SAR) was performed, which focused mainly on the combination of scaffold changes and revealed the key chemical features for potent anti-CHIKV inhibition. Further, a thorough ADMET investigation of the compounds was carried out: the compounds were screened for their aqueous solubility, lipophilicity, their toxicity in CaCo-2 cells, and possible hERG channel interactions. Additionally, 55 analogues were assessed for their metabolic stability in human liver microsomes (HLMs), leading to a structure-metabolism relationship study (SMR). The compounds showed an excellent safety profile, favourable physicochemical characteristics, and the required metabolic stability. A cross-resistance study confirmed the viral capping machinery (nsP1) to be the viral target of these compounds. This study identified 31b and 34 as potent, safe, and stable lead compounds for further development as selective CHIKV inhibitors. Finally, the collected insight led to a successful scaffold hop (64b) for future antiviral research studies.
Collapse
Affiliation(s)
- Verena Battisti
- Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Vienna, Josef-Holaubek-Platz 2, A-1090, Vienna, Austria.
| | - Julia Moesslacher
- Department of Pharmacy, University of Innsbruck, Innrain 80/82, A-6020, Innsbruck, Austria
| | - Rana Abdelnabi
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium
| | - Pieter Leyssen
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium
| | - Ana Lucia Rosales Rosas
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium
| | - Lana Langendries
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium
| | - Mohammed Aufy
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Josef-Holaubek-Platz 2, A-1090, Vienna, Austria
| | - Christian Studenik
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Josef-Holaubek-Platz 2, A-1090, Vienna, Austria
| | - Jadel M Kratz
- Department of Pharmaceutical Sciences, Division of Pharmacognosy, University of Vienna, Josef-Holaubek-Platz 2, A-1090, Vienna, Austria
| | - Judith M Rollinger
- Department of Pharmaceutical Sciences, Division of Pharmacognosy, University of Vienna, Josef-Holaubek-Platz 2, A-1090, Vienna, Austria
| | - Gerhard Puerstinger
- Department of Pharmacy, University of Innsbruck, Innrain 80/82, A-6020, Innsbruck, Austria
| | - Johan Neyts
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium
| | - Leen Delang
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium
| | - Ernst Urban
- Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Vienna, Josef-Holaubek-Platz 2, A-1090, Vienna, Austria
| | - Thierry Langer
- Department of Pharmaceutical Sciences, Pharmaceutical Chemistry Division, University of Vienna, Josef-Holaubek-Platz 2, A-1090, Vienna, Austria.
| |
Collapse
|
25
|
Kuno G. Mechanisms of Yellow Fever Transmission: Gleaning the Overlooked Records of Importance and Identifying Problems, Puzzles, Serious Issues, Surprises and Research Questions. Viruses 2024; 16:84. [PMID: 38257784 PMCID: PMC10820296 DOI: 10.3390/v16010084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/12/2023] [Accepted: 12/29/2023] [Indexed: 01/24/2024] Open
Abstract
In viral disease research, few diseases can compete with yellow fever for the volume of literature, historical significance, richness of the topics and the amount of strong interest among both scientists and laypersons. While the major foci of viral disease research shifted to other more pressing new diseases in recent decades, many critically important basic tasks still remain unfinished for yellow fever. Some of the examples include the mechanisms of transmission, the process leading to outbreak occurrence, environmental factors, dispersal, and viral persistence in nature. In this review, these subjects are analyzed in depth, based on information not only in old but in modern literatures, to fill in blanks and to update the current understanding on these topics. As a result, many valuable facts, ideas, and other types of information that complement the present knowledge were discovered. Very serious questions about the validity of the arbovirus concept and some research practices were also identified. The characteristics of YFV and its pattern of transmission that make this virus unique among viruses transmitted by Ae. aegypti were also explored. Another emphasis was identification of research questions. The discovery of a few historical surprises was an unexpected benefit.
Collapse
Affiliation(s)
- Goro Kuno
- Formerly at the Division of Vector-Borne Infectious Diseases, Centers for Disease Control and Prevention, Fort Collins, CO 80521, USA
| |
Collapse
|
26
|
Chitre SD, Crews CM, Tessema MT, Plėštytė-Būtienė I, Coffee M, Richardson ET. The impact of anthropogenic climate change on pediatric viral diseases. Pediatr Res 2024; 95:496-507. [PMID: 38057578 PMCID: PMC10872406 DOI: 10.1038/s41390-023-02929-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/12/2023] [Accepted: 11/16/2023] [Indexed: 12/08/2023]
Abstract
The adverse effects of climate change on human health are unfolding in real time. Environmental fragmentation is amplifying spillover of viruses from wildlife to humans. Increasing temperatures are expanding mosquito and tick habitats, introducing vector-borne viruses into immunologically susceptible populations. More frequent flooding is spreading water-borne viral pathogens, while prolonged droughts reduce regional capacity to prevent and respond to disease outbreaks with adequate water, sanitation, and hygiene resources. Worsening air quality and altered transmission seasons due to an increasingly volatile climate may exacerbate the impacts of respiratory viruses. Furthermore, both extreme weather events and long-term climate variation are causing the destruction of health systems and large-scale migrations, reshaping health care delivery in the face of an evolving global burden of viral disease. Because of their immunological immaturity, differences in physiology (e.g., size), dependence on caregivers, and behavioral traits, children are particularly vulnerable to climate change. This investigation into the unique pediatric viral threats posed by an increasingly inhospitable world elucidates potential avenues of targeted programming and uncovers future research questions to effect equitable, actionable change. IMPACT: A review of the effects of climate change on viral threats to pediatric health, including zoonotic, vector-borne, water-borne, and respiratory viruses, as well as distal threats related to climate-induced migration and health systems. A unique focus on viruses offers a more in-depth look at the effect of climate change on vector competence, viral particle survival, co-morbidities, and host behavior. An examination of children as a particularly vulnerable population provokes programming tailored to their unique set of vulnerabilities and encourages reflection on equitable climate adaptation frameworks.
Collapse
Affiliation(s)
- Smit D Chitre
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, MA, USA
| | - Cecilia M Crews
- Heilbrunn Department of Population & Family Health, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Mesfin Teklu Tessema
- Heilbrunn Department of Population & Family Health, Columbia University Mailman School of Public Health, New York, NY, USA.
- International Rescue Committee, New York, NY, USA.
| | | | - Megan Coffee
- Heilbrunn Department of Population & Family Health, Columbia University Mailman School of Public Health, New York, NY, USA
- International Rescue Committee, New York, NY, USA
- New York University Grossman School of Medicine, New York, NY, USA
| | - Eugene T Richardson
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
27
|
Rocha DCP, Sisnande T, Gavino-Leopoldino D, Guimarães-Andrade IP, Cruz FF, Assunção-Miranda I, Mendonça SC, Leitão GG, Simas RC, Mohana-Borges R, Leitão SG, Allonso D. Antiviral, Cytoprotective, and Anti-Inflammatory Effect of Ampelozizyphus amazonicus Ducke Ethanolic Wood Extract on Chikungunya Virus Infection. Viruses 2023; 15:2232. [PMID: 38005909 PMCID: PMC10674702 DOI: 10.3390/v15112232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Chikungunya fever, a debilitating disease caused by Chikungunya virus (CHIKV), is characterized by a high fever of sudden onset and an intense arthralgia that impairs individual regular activities. Although most symptoms are self-limited, long-term persistent arthralgia is observed in 30-40% of infected individuals. Currently, there is no vaccine or specific treatment against CHIKV infection, so there is an urgent need for the discovery of new therapeutic options for CHIKF chronic cases. This present study aims to test the antiviral, cytoprotective, and anti-inflammatory activities of an ethanol extract (FF72) from Ampelozizyphus amazonicus Ducke wood, chemically characterized using mass spectrometry, which indicated the major presence of dammarane-type triterpenoid saponins. The major saponin in the extract, with a deprotonated molecule ion m/z 897 [M-H]-, was tentatively assigned as a jujubogenin triglycoside, a dammarane-type triterpenoid saponin. Treatment with FF72 resulted in a significant reduction in both virus replication and the production of infective virions in BHK-21-infected cells. The viability of infected cells was assessed using an MTT, and the result indicated that FF72 treatment was able to revert the toxicity mediated by CHIKV infection. In addition, FF72 had a direct effect on CHIKV, since the infectivity was completely abolished in the presence of the extract. FF72 treatment also reduced the expression of the major pro-inflammatory mediators overexpressed during CHIKV infection, such as IL-1β, IL-6, IL-8, and MCP-1. Overall, the present study elucidates the potential of FF72 to become a promising candidate of herbal medicine for alphaviruses infections.
Collapse
Affiliation(s)
- Daniele C. P. Rocha
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (T.S.); (R.M.-B.)
| | - Tháyna Sisnande
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (T.S.); (R.M.-B.)
| | - Daniel Gavino-Leopoldino
- Laboratório de Resposta Celular à Infecções Virais, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (D.G.-L.); (I.P.G.-A.); (I.A.-M.)
| | - Iris Paula Guimarães-Andrade
- Laboratório de Resposta Celular à Infecções Virais, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (D.G.-L.); (I.P.G.-A.); (I.A.-M.)
| | - Fernanda F. Cruz
- Laboratório de Investigação Pulmonar, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
| | - Iranaia Assunção-Miranda
- Laboratório de Resposta Celular à Infecções Virais, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (D.G.-L.); (I.P.G.-A.); (I.A.-M.)
| | - Simony C. Mendonça
- Instituto de Pesquisas de Produtos Naturais, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (S.C.M.); (G.G.L.)
- Departamento de Produtos Naturais e Alimentos, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
| | - Gilda Guimarães Leitão
- Instituto de Pesquisas de Produtos Naturais, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (S.C.M.); (G.G.L.)
| | - Rosineide Costa Simas
- Faculdade de Química, Escola de Engenharia, Universidade Presbiteriana Mackenzie, São Paulo 01302-907, SP, Brazil;
| | - Ronaldo Mohana-Borges
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (T.S.); (R.M.-B.)
| | - Suzana Guimarães Leitão
- Departamento de Produtos Naturais e Alimentos, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
| | - Diego Allonso
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
| |
Collapse
|
28
|
Atella MO, Carvalho AS, Da Poian AT. Role of macrophages in the onset, maintenance, or control of arthritis caused by alphaviruses. Exp Biol Med (Maywood) 2023; 248:2039-2044. [PMID: 38058027 PMCID: PMC10800133 DOI: 10.1177/15353702231214261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023] Open
Abstract
Arthritogenic alphaviruses are mosquito-borne viruses that cause a debilitating rheumatic disease characterized by fever, headache, rash, myalgia, and polyarthralgia with the potential to evolve into a severe and very prolonged illness. Although these viruses have been geographically restricted by vector hosts and reservoirs, recent epidemics have revealed the risks of their spread worldwide. In this review, we aim to discuss the protective and pathological roles of macrophages during the development of arthritis caused by alphaviruses. The progression to the chronic phase of the disease is related to the extension of viral replication and the maintenance of articular inflammation, in which the cellular infiltrate is predominantly composed of macrophages. We explore the possible implications of macrophage polarization to M1/M2 activation phenotypes, drawing a parallel between alphavirus arthritis and rheumatoid arthritis (RA), a chronic inflammatory disease that also affects articular tissues. In RA, it is well established that M1 macrophages contribute to tissue damage and inflammation, while M2 macrophages have a role in cartilage repair, so modulating the M1/M2 macrophage ratio is being considered as a strategy in the treatment of this disease. In the case of alphavirus-induced arthritis, the picture is more complex, as proinflammatory factors derived from M1 macrophages contribute to the antiviral response but cause tissue damage, while M2 macrophages may contribute to tissue repair but impair viral clearance.
Collapse
Affiliation(s)
| | | | - Andrea T Da Poian
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| |
Collapse
|
29
|
Nguyen TV, Ngwe Tun MM, Cao MT, Dao HM, Luong CQ, Huynh TKL, Nguyen TTT, Hoang TND, Morita K, Le TQM, Pham QD, Takamatsu Y, Hasebe F. Serological and Molecular Epidemiology of Chikungunya Virus Infection in Vietnam, 2017-2019. Viruses 2023; 15:2065. [PMID: 37896842 PMCID: PMC10611313 DOI: 10.3390/v15102065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 09/30/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Chikungunya fever is an acute febrile illness caused by the chikungunya virus (CHIKV), which is transmitted by Aedes mosquitoes. Since 1965, only a few studies with limited scope have been conducted on CHIKV in Vietnam. Thus, this study aimed to determine the seroprevalence and molecular epidemiology of CHIKV infection among febrile patients in Vietnam from 2017 to 2019. A total of 1063 serum samples from 31 provinces were collected and tested for anti-CHIKV IgM and IgG ELISA. The 50% focus reduction neutralization test (FRNT50) was used to confirm CHIKV-neutralizing antibodies. Quantitative real-time RT-PCR (RT-qPCR) was performed to confirm the presence of the CHIKV genome. The results showed that 15.9% (169/1063) of the patients had anti-CHIKV IgM antibodies, 20.1% (214/1063) had anti-CHIKV IgG antibodies, 10.4% (111/1063) had CHIKV-neutralizing antibodies, and 27.7% (130/469) of the samples were positive in RT-qPCR analysis. The E1 CHIKV genome sequences were detected among the positive RT-qPCR samples. Our identified sequences belonged to the East/Central/South/African (ECSA) genotype, which has been prevalent in Vietnam previously, suggesting CHIKV has been maintained and is endemic in Vietnam. This study demonstrates a high prevalence of CHIKV infection in Vietnam and calls for an annual surveillance program to understand its impact.
Collapse
Affiliation(s)
- Thanh Vu Nguyen
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan; (T.V.N.); (K.M.)
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (M.T.C.); (H.M.D.); (C.Q.L.); (T.K.L.H.); (T.T.T.N.); (T.N.D.H.); (Q.D.P.)
| | - Mya Myat Ngwe Tun
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan; (T.V.N.); (K.M.)
- DEJIMA Infectious Disease Research Alliance, Nagasaki University, Nagasaki 852-8523, Japan
- Center for Vaccines and Therapeutic Antibodies for Emerging Infectious Diseases, Shimane University, Izumo 690-8504, Japan
| | - Minh Thang Cao
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (M.T.C.); (H.M.D.); (C.Q.L.); (T.K.L.H.); (T.T.T.N.); (T.N.D.H.); (Q.D.P.)
| | - Huy Manh Dao
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (M.T.C.); (H.M.D.); (C.Q.L.); (T.K.L.H.); (T.T.T.N.); (T.N.D.H.); (Q.D.P.)
| | - Chan Quang Luong
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (M.T.C.); (H.M.D.); (C.Q.L.); (T.K.L.H.); (T.T.T.N.); (T.N.D.H.); (Q.D.P.)
| | - Thi Kim Loan Huynh
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (M.T.C.); (H.M.D.); (C.Q.L.); (T.K.L.H.); (T.T.T.N.); (T.N.D.H.); (Q.D.P.)
| | - Thi Thanh Thuong Nguyen
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (M.T.C.); (H.M.D.); (C.Q.L.); (T.K.L.H.); (T.T.T.N.); (T.N.D.H.); (Q.D.P.)
| | - Thi Nhu Dao Hoang
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (M.T.C.); (H.M.D.); (C.Q.L.); (T.K.L.H.); (T.T.T.N.); (T.N.D.H.); (Q.D.P.)
| | - Kouichi Morita
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan; (T.V.N.); (K.M.)
- DEJIMA Infectious Disease Research Alliance, Nagasaki University, Nagasaki 852-8523, Japan
| | - Thi Quynh Mai Le
- National Institute of Hygiene and Epidemiology, Hanoi 100000, Vietnam;
| | - Quang Duy Pham
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (M.T.C.); (H.M.D.); (C.Q.L.); (T.K.L.H.); (T.T.T.N.); (T.N.D.H.); (Q.D.P.)
| | - Yuki Takamatsu
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan; (T.V.N.); (K.M.)
- DEJIMA Infectious Disease Research Alliance, Nagasaki University, Nagasaki 852-8523, Japan
| | - Futoshi Hasebe
- Vietnam Research Station, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan
| |
Collapse
|
30
|
Kayange N, Hau DK, Pain K, Mshana SE, Peck R, Gehring S, Groendahl B, Koliopoulos P, Revocatus B, Msaki EB, Malande O. Seroprevalence of Dengue and Chikungunya Virus Infections in Children Living in Sub-Saharan Africa: Systematic Review and Meta-Analysis. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1662. [PMID: 37892325 PMCID: PMC10605353 DOI: 10.3390/children10101662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/23/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023]
Abstract
Dengue and chikungunya viruses are frequent causes of malarial-like febrile illness in children. The rapid increase in virus transmission by mosquitoes is a global health concern. This is the first systematic review and meta-analysis of the childhood prevalence of dengue and chikungunya in Sub-Saharan Africa (SSA). A comprehensive search of the MEDLINE (Ovid), Embase (Ovid), and Cochrane Library (Wiley) databases was conducted on 28 June 2019, and updated on 12 February 2022. The search strategy was designed to retrieve all articles pertaining to arboviruses in SSA children using both controlled vocabulary and keywords. The pooled (weighted) proportion of dengue and chikungunya was estimated using a random effect model. The overall pooled prevalence of dengue and chikungunya in SSA children was estimated to be 16% and 7%, respectively. Prevalence was slightly lower during the period 2010-2020 compared to 2000-2009. The study design varied depending on the healthcare facility reporting the disease outbreak. Importantly, laboratory methods used to detect arbovirus infections differed. The present review documents the prevalence of dengue and chikungunya in pediatric patients throughout SSA. The results provide unprecedented insight into the transmission of dengue and chikungunya viruses among these children and highlight the need for enhanced surveillance and controlled methodology.
Collapse
Affiliation(s)
- Neema Kayange
- Department of Pediatrics, Bugando Medical Centre, Weill Bugando School of Medicine, Catholic University of Health and Allied Sciences, Mwanza P.O. Box 1464, Tanzania;
| | - Duncan K Hau
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA;
| | - Kevin Pain
- Samuel J. Wood Library and C.V. Starr Biomedical Information Center, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA;
| | - Stephen E Mshana
- Department of Microbiology and Immunology, Weill Bugando School of Medicine, Catholic University of Health and Allied Sciences, Mwanza P.O. Box 1464, Tanzania;
| | - Robert Peck
- Department of Pediatrics, Bugando Medical Centre, Weill Bugando School of Medicine, Catholic University of Health and Allied Sciences, Mwanza P.O. Box 1464, Tanzania;
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA;
- Center for Global Health, Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Stephan Gehring
- Department of Pediatrics, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (S.G.); (B.G.); (P.K.)
| | - Britta Groendahl
- Department of Pediatrics, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (S.G.); (B.G.); (P.K.)
| | - Philip Koliopoulos
- Department of Pediatrics, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany; (S.G.); (B.G.); (P.K.)
| | - Baraka Revocatus
- Department of Data and Statistics, Bugando Medical Centre, Mwanza P.O. Box 1370, Tanzania;
| | - Evarist B Msaki
- Department of Epidemiology and Biostatistics, Bugando Medical Centre, Mwanza P.O. Box 1370, Tanzania;
| | - Ombeva Malande
- East Africa Centre for Vaccines and Immunization (ECAVI), Kampala P.O. Box 3040, Uganda;
- Department of Public Health Phamarmacy, Sefako Makgatho Health Sciences University, Pretoria P.O. Box 60, South Africa
- Department of Paediatrics & Child Health, Makerere University, Kampala P.O. Box 7072, Uganda
- Department of Public Health, UNICAF University, Lusaka P.O. Box 20842, Zambia
| |
Collapse
|
31
|
Tretyakova I, Joh J, Lukashevich IS, Alejandro B, Gearon M, Chung D, Pushko P. Live-Attenuated CHIKV Vaccine with Rearranged Genome Replicates in vitro and Induces Immune Response in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.16.558061. [PMID: 37745520 PMCID: PMC10516039 DOI: 10.1101/2023.09.16.558061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Chikungunya fever virus (CHIKV) is a mosquito-borne alphavirus that causes wide-spread human infections and epidemics in Asia, Africa and recently, in the Americas. There is no approved vaccine and CHIKV is considered a priority pathogen by CEPI and WHO. Previously, we developed immunization DNA (iDNA) plasmid capable of launching live-attenuated CHIKV vaccine in vivo . Here we report the use of CHIKV iDNA plasmid to prepare a novel, live-attenuated CHIKV vaccine V5040 with rearranged RNA genome for improved safety. In V5040, genomic RNA was rearranged to encode capsid gene downstream from the glycoprotein genes. To secure safety profile, attenuated mutations derived from experimental CHIKV 181/25 vaccine were also engineered into E2 gene of V5040. The DNA copy of rearranged CHIKV genomic RNA with attenuated mutations was cloned into iDNA plasmid pMG5040 downstream from the CMV promoter. After transfection in vitro, pMG5040 launched replication of V5040 virus with rearranged genome and attenuating E2 mutations. Furthermore, V5040 virus was evaluated in experimental murine models for safety and immunogenicity. Vaccination with V5040 virus subcutaneously resulted in elicitation of CHIKV-specific, virus-neutralizing antibodies. The results warrant further evaluation of V5040 virus with rearranged genome as a novel live-attenuated vaccine for CHIKV.
Collapse
|
32
|
Dommar CJ, López L, Paul R, Rodó X. The 2013 Chikungunya outbreak in the Caribbean was structured by the network of cultural relationships among islands. ROYAL SOCIETY OPEN SCIENCE 2023; 10:230909. [PMID: 37711149 PMCID: PMC10498052 DOI: 10.1098/rsos.230909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023]
Abstract
In 2013, the Caribbean underwent an unprecedented epidemic of Chikungunya that affected 29 islands and mainland territories throughout the Caribbean in the first six months. Analysing the spread of the epidemic among the Caribbean islands, we show that the initial patterns of the epidemic can be explained by a network model based on the flight connections among islands. The network does not follow a random graph model and its topology is likely the product of geo-political relationships that generate increased connectedness among locations sharing the same language. Therefore, the infection propagated preferentially among islands that belong to the same cultural domain, irrespective of their human and vector population densities. Importantly, the flight network topology was also a more important determinant of the disease dynamics than the actual volume of traffic. Finally, the severity of the epidemic was found to depend, in the first instance, on which island was initially infected. This investigation shows how a simple epidemic model coupled with an appropriate human mobility model can reproduce the observed epidemiological dynamics. Also, it sheds light on the design of interventions in the face of the emergence of infections in similar settings of naive subpopulations loosely interconnected by host movement. This study delves into the feasibility of developing models to anticipate the emergence of vector-borne infections, showing the importance of network topology, bringing valuable methods for public health officials when planning control policies. Significance statement: The study shows how a simple epidemic model associated with an appropriate human mobility model can reproduce the observed epidemiological dynamics of the 2014 Chikungunya epidemic in the Caribbean region. This model sheds light on the design of interventions in the face of the emergence of infections in similar settings of naive subpopulations loosely interconnected by the host.
Collapse
Affiliation(s)
- Carlos J. Dommar
- Theoretical and Computational Ecology Group, Centre d’Estudis Avanßats de Blanes CSIC-CEAB, Blanes 17300, Spain
- CLIMA Climate and Health Program, ISGlobal, Barcelona 08003, Spain
| | - Leonardo López
- CLIMA Climate and Health Program, ISGlobal, Barcelona 08003, Spain
| | - Richard Paul
- Ecology and Emergence of Arthropod-borne Pathogens unit, Institut Pasteur, Université Paris-Cité, Centre National de Recherche Scientifique (CNRS) UMR 2000, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE) USC 1510, 75015 Paris, France
- Centre National de la Recherche Scientifique (CNRS), Génomique évolutive, modélisation et santé UMR 2000, 75724 Paris Cedex 15, France
| | - Xavier Rodó
- CLIMA Climate and Health Program, ISGlobal, Barcelona 08003, Spain
- ICREA, Barcelona, 08010 Catalonia, Spain
| |
Collapse
|
33
|
Obame-Nkoghe J, Roiz D, Ngangue MF, Costantini C, Rahola N, Jiolle D, Lehmann D, Makaga L, Ayala D, Kengne P, Paupy C. Towards the invasion of wild and rural forested areas in Gabon (Central Africa) by the Asian tiger mosquito Aedes albopictus: Potential risks from the one health perspective. PLoS Negl Trop Dis 2023; 17:e0011501. [PMID: 37585443 PMCID: PMC10461836 DOI: 10.1371/journal.pntd.0011501] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 08/28/2023] [Accepted: 07/05/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND Since its first record in urban areas of Central-Africa in the 2000s, the invasive mosquito, Aedes albopictus, has spread throughout the region, including in remote villages in forested areas, causing outbreaks of Aedes-borne diseases, such as dengue and chikungunya. Such invasion might enhance Ae. albopictus interactions with wild animals in forest ecosystems and favor the spillover of zoonotic arboviruses to humans. The aim of this study was to monitor Ae. albopictus spread in the wildlife reserve of La Lopé National Park (Gabon), and evaluate the magnitude of the rainforest ecosystem colonization. METHODOLOGY From 2014 to 2018, we used ovitraps, larval surveys, BG-Sentinel traps, and human landing catches along an anthropization gradient from La Lopé village to the natural forest in the Park. CONCLUSIONS We detected Ae. albopictus in gallery forest up to 15 km away from La Lopé village. However, Ae. albopictus was significantly more abundant at anthropogenic sites than in less anthropized areas. The number of eggs laid by Ae. albopictus decreased progressively with the distance from the forest fringe up to 200m inside the forest. Our results suggested that in forest ecosystems, high Ae. albopictus density is mainly observed at interfaces between anthropized and natural forested environments. Additionally, our data suggested that Ae. albopictus may act as a bridge vector of zoonotic pathogens between wild and anthropogenic compartments.
Collapse
Affiliation(s)
- Judicaël Obame-Nkoghe
- Laboratoire de Biologie Moléculaire et Cellulaire, Département de Biologie, Université des Sciences et Techniques de Masuku, Franceville, Gabon
- Unité de Recherche en Écologie de la Santé, Centre Interdisciplinaire de Recherches Médicales de Franceville, Franceville, Gabon
| | - David Roiz
- MIVEGEC, Univ. Montpellier, CNRS, IRD, Montpellier, France
| | - Marc-Flaubert Ngangue
- Unité de Recherche en Écologie de la Santé, Centre Interdisciplinaire de Recherches Médicales de Franceville, Franceville, Gabon
- Agence Nationale des Parcs Nationaux, Quartier Haut de Gué Gué, Libreville, Gabon
| | | | - Nil Rahola
- MIVEGEC, Univ. Montpellier, CNRS, IRD, Montpellier, France
| | - Davy Jiolle
- MIVEGEC, Univ. Montpellier, CNRS, IRD, Montpellier, France
| | - David Lehmann
- Agence Nationale des Parcs Nationaux, Quartier Haut de Gué Gué, Libreville, Gabon
- Biological and Environmental Sciences, University of Stirling, Stirling, United Kingdom
| | - Loïc Makaga
- Agence Nationale des Parcs Nationaux, Quartier Haut de Gué Gué, Libreville, Gabon
| | - Diego Ayala
- MIVEGEC, Univ. Montpellier, CNRS, IRD, Montpellier, France
| | - Pierre Kengne
- Unité de Recherche en Écologie de la Santé, Centre Interdisciplinaire de Recherches Médicales de Franceville, Franceville, Gabon
- MIVEGEC, Univ. Montpellier, CNRS, IRD, Montpellier, France
| | | |
Collapse
|
34
|
Abdoullah B, Durand GA, Basco LK, El Bara A, Bollahi MA, Bosio L, Geulen M, Briolant S, Boukhary AOMS. Seroprevalence of Alphaviruses ( Togaviridae) among Urban Population in Nouakchott, Mauritania, West Africa. Viruses 2023; 15:1588. [PMID: 37515274 PMCID: PMC10385508 DOI: 10.3390/v15071588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
The presence of alphaviruses, such as chikungunya virus (CHIKV), has never been reported in Mauritania. We assessed the seroprevalence of CHIKV among Nouakchott residents. A cross-sectional study involving 1300 non-febrile patients consulting at the Nouakchott hospital center was conducted between January and June 2021. The presence of anti-CHIKV IgG and neutralizing antibodies against CHIKV, O'nyong-nyong virus (ONNV), and Semliki Forest virus (SFV) was determined by an enzyme-linked immunosorbent assay (ELISA) and a serum neutralization test, respectively, and the associated risk factors were investigated. Of the 1300 study participants, serological evidence of previous exposure to CHIKV was observed in 37 individuals (2.8%). Sex, age, reported use of repellants, and bed net ownership and usage were not associated with CHIKV seropositivity. Our results showed the co-circulation of two other alphaviruses, ONNV and SFV, in Nouakchott in 30 (2.3%) individuals. This is the first study that documents the co-circulation of CHIKV, ONNV, and SFV in Mauritania, albeit at low prevalence. Surveillance and routine testing for alphaviruses and other arboviruses in symptomatic patients should be implemented in health facilities to assess the health burden associated with these viruses. Efforts should also be made to strengthen the vector control measures.
Collapse
Affiliation(s)
- Bedia Abdoullah
- Unité de Recherche Génomes et Milieux (GEMI), Université de Nouakchott, Nouveau Campus Universitaire, Nouakchott BP 5026, Mauritania
| | - Guillaume André Durand
- National Reference Center for Arboviruses, National Institute of Health and Medical Research (Inserm) and French Armed Forces Biomedical Research Institute (IRBA), 13005 Marseille, France
- Unité des Virus Émergents (UVE: Aix-Marseille Univ-IRD 190-Inserm 1207), 13005 Marseille, France
| | - Leonardo K Basco
- Aix Marseille Univ, IRD, AP-HM, SSA, VITROME, 13005 Marseille, France
- IHU-Méditerranée Infection, 13005 Marseille, France
| | - Ahmed El Bara
- Institut National de Recherche en Santé Publique, Nouakchott BP 695, Mauritania
| | | | - Laurent Bosio
- National Reference Center for Arboviruses, National Institute of Health and Medical Research (Inserm) and French Armed Forces Biomedical Research Institute (IRBA), 13005 Marseille, France
- Unité des Virus Émergents (UVE: Aix-Marseille Univ-IRD 190-Inserm 1207), 13005 Marseille, France
| | - Manon Geulen
- National Reference Center for Arboviruses, National Institute of Health and Medical Research (Inserm) and French Armed Forces Biomedical Research Institute (IRBA), 13005 Marseille, France
- Unité des Virus Émergents (UVE: Aix-Marseille Univ-IRD 190-Inserm 1207), 13005 Marseille, France
| | - Sébastien Briolant
- Aix Marseille Univ, IRD, AP-HM, SSA, VITROME, 13005 Marseille, France
- IHU-Méditerranée Infection, 13005 Marseille, France
- Unité de Parasitologie Entomologie, Département de Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées (IRBA), 13005 Marseille, France
| | - Ali Ould Mohamed Salem Boukhary
- Unité de Recherche Génomes et Milieux (GEMI), Université de Nouakchott, Nouveau Campus Universitaire, Nouakchott BP 5026, Mauritania
| |
Collapse
|
35
|
Amaral JK, Taylor PC, Schoen RT. Brazil at the Center of Chikungunya Outbreaks. J Glob Infect Dis 2023; 15:131-132. [PMID: 37800080 PMCID: PMC10549908 DOI: 10.4103/jgid.jgid_21_23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/29/2023] [Indexed: 10/07/2023] Open
Affiliation(s)
- J. Kennedy Amaral
- Department of Rheumatology, Institute of Diagnostic Medicine of Cariri, Juazeiro do Norte, Ceará, Brazil
| | - Peter C. Taylor
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Robert T. Schoen
- Clinical Professor of Medicine, Section of Rheumatology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
36
|
Barker D, Han X, Wang E, Dagley A, Anderson DM, Jha A, Weaver SC, Julander J, Nykiforuk C, Kodihalli S. Equine Polyclonal Antibodies Prevent Acute Chikungunya Virus Infection in Mice. Viruses 2023; 15:1479. [PMID: 37515166 PMCID: PMC10384969 DOI: 10.3390/v15071479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted pathogen that causes chikungunya disease (CHIK); the disease is characterized by fever, muscle ache, rash, and arthralgia. This arthralgia can be debilitating and long-lasting, seriously impacting quality of life for years. Currently, there is no specific therapy available for CHIKV infection. We have developed a despeciated equine polyclonal antibody (CHIKV-EIG) treatment against CHIKV and evaluated its protective efficacy in mouse models of CHIKV infection. In immunocompromised (IFNAR-/-) mice infected with CHIKV, daily treatment for five consecutive days with CHIKV-EIG administered at 100 mg/kg starting on the day of infection prevented mortality, reduced viremia, and improved clinical condition as measured by body weight loss. These beneficial effects were seen even when treatment was delayed to 1 day after infection. In immunocompetent mice, CHIKV-EIG treatment reduced virus induced arthritis (including footpad swelling), arthralgia-associated cytokines, viremia, and tissue virus loads in a dose-dependent fashion. Collectively, these results suggest that CHIKV-EIG is effective at preventing CHIK and could be a viable candidate for further development as a treatment for human disease.
Collapse
Affiliation(s)
- Douglas Barker
- Emergent BioSolutions Canada Inc., Winnipeg, MB R3T 5Y3, Canada
| | - Xiaobing Han
- Emergent BioSolutions Canada Inc., Winnipeg, MB R3T 5Y3, Canada
| | - Eryu Wang
- Institute for Human Infections and Immunity, Department of Microbiology and Immunology, University of Texas Medical Branch Galveston, Galveston, TX 77555, USA
| | - Ashley Dagley
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | | | - Aruni Jha
- Emergent BioSolutions Canada Inc., Winnipeg, MB R3T 5Y3, Canada
| | - Scott C Weaver
- Institute for Human Infections and Immunity, Department of Microbiology and Immunology, University of Texas Medical Branch Galveston, Galveston, TX 77555, USA
| | - Justin Julander
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Cory Nykiforuk
- Emergent BioSolutions Canada Inc., Winnipeg, MB R3T 5Y3, Canada
| | | |
Collapse
|
37
|
Stephenson KE. Live-attenuated Chikungunya vaccine: a possible new era. Lancet 2023; 401:2090-2091. [PMID: 37321234 DOI: 10.1016/s0140-6736(23)01170-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/17/2023]
Affiliation(s)
- Kathryn E Stephenson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA.
| |
Collapse
|
38
|
Rao S, Abeyratne E, Freitas JR, Yang C, Tharmarajah K, Mostafavi H, Liu X, Zaman M, Mahalingam S, Zaid A, Taylor A. A booster regime of liposome-delivered live-attenuated CHIKV vaccine RNA genome protects against chikungunya virus disease in mice. Vaccine 2023; 41:3976-3988. [PMID: 37230889 DOI: 10.1016/j.vaccine.2023.05.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/27/2023]
Abstract
Mosquito-transmitted chikungunya virus (CHIKV) is the causal pathogen of CHIKV disease and is responsible for global epidemics of arthritic disease. CHIKV infection can lead to severe chronic and debilitating arthralgia, significantly impacting patient mobility and quality of life. Our previous studies have shown a live-attenuated CHIKV vaccine candidate, CHIKV-NoLS, to be effective in protecting against CHIKV disease in mice vaccinated with one dose. Further studies have demonstrated the value of a liposome RNA delivery system to deliver the RNA genome of CHIKV-NoLS directly in vivo, promoting de novo production of live-attenuated vaccine particles in vaccinated hosts. This system, designed to bypass live-attenuated vaccine production bottlenecks, uses CAF01 liposomes. However, one dose of CHIKV-NoLS CAF01 failed to provide systemic protection against CHIKV challenge in mice, with low levels of CHIKV-specific antibodies. Here we describe CHIKV-NoLS CAF01 booster vaccination regimes designed to increase vaccine efficacy. C57BL/6 mice were vaccinated with three doses of CHIKV-NoLS CAF01 either intramuscularly or subcutaneously. CHIKV-NoLS CAF01 vaccinated mice developed a systemic immune response against CHIKV that shared similarity to vaccination with CHIKV-NoLS, including high levels of CHIKV-specific neutralising antibodies in subcutaneously inoculated mice. CHIKV-NoLS CAF01 vaccinated mice were protected against disease signs and musculoskeletal inflammation when challenged with CHIKV. Mice given one dose of live-attenuated CHIKV-NoLS developed a long lasting protective immune response for up to 71 days. A clinically relevant CHIKV-NoLS CAF01 booster regime can overcome the challenges faced by our previous one dose strategy and provide systemic protection against CHIKV disease.
Collapse
Affiliation(s)
- Shambhavi Rao
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Eranga Abeyratne
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Joseph R Freitas
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Chenying Yang
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Kothila Tharmarajah
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Helen Mostafavi
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Xiang Liu
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Mehfuz Zaman
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, 4222 Queensland, Australia
| | - Suresh Mahalingam
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Ali Zaid
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia
| | - Adam Taylor
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD 4215, Australia; School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Southport, 4215 Queensland, Australia; Global Virus Network (GVN) Centre for Excellence in Arboviruses, Australia.
| |
Collapse
|
39
|
Simon F, Caumes E, Jelinek T, Lopez-Velez R, Steffen R, Chen LH. Chikungunya: risks for travellers. J Travel Med 2023; 30:taad008. [PMID: 36648431 PMCID: PMC10075059 DOI: 10.1093/jtm/taad008] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023]
Abstract
RATIONALE FOR REVIEW Chikungunya outbreaks continue to occur, with changing epidemiology. Awareness about chikungunya is low both among the at-risk travellers and healthcare professionals, which can result in underdiagnosis and underreporting. This review aims to improve awareness among healthcare professionals regarding the risks of chikungunya for travellers. KEY FINDINGS Chikungunya virus transmission to humans occurs mainly via daytime-active mosquitoes, Aedes aegypti and Aedes albopictus. The areas where these mosquitoes live is continuously expanding, partly due to climate changes. Chikungunya is characterized by an acute onset of fever with joint pain. These symptoms generally resolve within 1-3 weeks, but at least one-third of the patients suffer from debilitating rheumatologic symptoms for months to years. Large outbreaks in changing regions of the world since the turn of the 21st century (e.g. Caribbean, La Réunion; currently Brazil, India) have resulted in growing numbers of travellers importing chikungunya, mainly to Europe and North America. Viremic travellers with chikungunya infection have seeded chikungunya clusters (France, United States of America) and outbreaks (Italy in 2007 and 2017) in non-endemic countries where Ae. albopictus mosquitoes are present. Community preventive measures are important to prevent disease transmission by mosquitoes. Individual preventive options are limited to personal protection measures against mosquito bites, particularly the daytime-active mosquitos that transmit the chikungunya virus. Candidate vaccines are on the horizon and regulatory authorities will need to assess environmental and host risk factors for persistent sequelae, such as obesity, age (over 40 years) and history of arthritis or inflammatory rheumatologic disease to determine which populations should be targeted for these chikungunya vaccines. CONCLUSIONS/RECOMMENDATIONS Travellers planning to visit destinations with active CHIKV circulation should be advised about the risk for chikungunya, prevention strategies, the disease manifestations, possible chronic rheumatologic sequelae and, if symptomatic, seek medical evaluation and report potential exposures.
Collapse
Affiliation(s)
- Fabrice Simon
- Service de Pathologie Infectieuse et Tropicale, Hôpital d’Instruction des Armées Laveran, Marseille, France
| | - Eric Caumes
- Centre de Diagnostic, Hôpital de l’Hôtel-Dieu, Paris, France
| | - Tomas Jelinek
- Berlin Centre for Travel and Tropical Medicine, Berlin, Germany
| | - Rogelio Lopez-Velez
- Ramón y Cajal Institute for Health Research (IRyCIS), Ramón y Cajal University Hospital, Madrid, Spain
| | - Robert Steffen
- Epidemiology, Biostatistics and Prevention Institute, WHO Collaborating Center on Travelers’ Health, University of Zurich, Zurich, Switzerland
- Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas School of Public Health, Houston, TX, USA
| | - Lin H Chen
- Division of Infectious Diseases and Travel Medicine, Mount Auburn Hospital, Cambridge, MA, USA
- Faculty of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
40
|
Novelo M, Dutra HLC, Metz HC, Jones MJ, Sigle LT, Frentiu FD, Allen SL, Chenoweth SF, McGraw EA. Dengue and chikungunya virus loads in the mosquito Aedes aegypti are determined by distinct genetic architectures. PLoS Pathog 2023; 19:e1011307. [PMID: 37043515 PMCID: PMC10124881 DOI: 10.1371/journal.ppat.1011307] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/24/2023] [Accepted: 03/19/2023] [Indexed: 04/13/2023] Open
Abstract
Aedes aegypti is the primary vector of the arboviruses dengue (DENV) and chikungunya (CHIKV). These viruses exhibit key differences in their vector interactions, the latter moving more quicky through the mosquito and triggering fewer standard antiviral pathways. As the global footprint of CHIKV continues to expand, we seek to better understand the mosquito's natural response to CHIKV-both to compare it to DENV:vector coevolutionary history and to identify potential targets in the mosquito for genetic modification. We used a modified full-sibling design to estimate the contribution of mosquito genetic variation to viral loads of both DENV and CHIKV. Heritabilities were significant, but higher for DENV (40%) than CHIKV (18%). Interestingly, there was no genetic correlation between DENV and CHIKV loads between siblings. These data suggest Ae. aegypti mosquitoes respond to the two viruses using distinct genetic mechanisms. We also examined genome-wide patterns of gene expression between High and Low CHIKV families representing the phenotypic extremes of viral load. Using RNAseq, we identified only two loci that consistently differentiated High and Low families: a long non-coding RNA that has been identified in mosquito screens post-infection and a distant member of a family of Salivary Gland Specific (SGS) genes. Interestingly, the latter gene is also associated with horizontal gene transfer between mosquitoes and the endosymbiotic bacterium Wolbachia. This work is the first to link the SGS gene to a mosquito phenotype. Understanding the molecular details of how this gene contributes to viral control in mosquitoes may, therefore, also shed light on its role in Wolbachia.
Collapse
Affiliation(s)
- Mario Novelo
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Heverton LC Dutra
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Hillery C. Metz
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Matthew J. Jones
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Leah T. Sigle
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Francesca D. Frentiu
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Herston, Queensland, Australia
| | - Scott L. Allen
- School of Biological Sciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Stephen F. Chenoweth
- School of Biological Sciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Elizabeth A. McGraw
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
41
|
Moreira TP, Sousa CDFD, Melo Costa VRD, Queiroz-Junior CM, Santos FM, Bonilha CS, Ésper LM, Nogueira ML, Cunha TM, Teixeira MM, Costa VV, de Souza DDG. Tumour necrosis factor plays a deleterious role in the pathogenesis of chikungunya virus infection. Immunology 2023; 168:444-458. [PMID: 36164989 DOI: 10.1111/imm.13583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 09/01/2022] [Indexed: 11/28/2022] Open
Abstract
Arthralgia is a hallmark of chikungunya virus (CHIKV) infection and can be very debilitating and associated with a robust local inflammatory response. Many pathophysiological aspects associated with the disease remain to be elucidated. Here, we describe a novel model of CHIKV infection in immunocompetent mice and evaluate the role of tumour necrosis factor in the pathogenesis of the disease. C57BL/6 wild type (WT) or TNF receptor 1 deficient (TNFR1-/- ) mice were inoculated with 1 × 106 PFU of CHIKV in the paw. Alternatively, etanercept was used to inhibit TNF in infected WT mice. Hypernociception, inflammatory and virological analysis were performed. Inoculation of CHIKV into WT mice induced persistent hypernociception. There was significant viral replication in target organs and local production of inflammatory mediators in early time-points after infection. CHIKV infection was associated with specific humoral IgM and IgG responses. In TNFR1-/- mice, there was a decrease in the hypernociception threshold, which was associated with a milder local inflammatory response in the paw but delayed viral clearance. Local or systemic treatment with etanercept reduced CHIKV-induced hypernociception. This is the first study to describe hypernociception, a clinical correlation of arthralgia, in immunocompetent mice infected with CHIKV. It also demonstrates the dual role of TNF in contributing to viral clearance but driving tissue damage and hypernociception. Inhibition of TNF may have therapeutic benefits but its role in viral clearance suggests that viral levels must be monitored in CHIKV-infected patients and that TNF inhibitors should ideally be used in combination with anti-viral drugs.
Collapse
Affiliation(s)
- Thaiane Pinto Moreira
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | - Franciele Martins Santos
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Caio Santos Bonilha
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Lísia Maria Ésper
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mauricio Lacerda Nogueira
- Department of Dermatological, Infectious and Parasitic Diseases, Medical School of São José do Rio Preto, São Paulo, São José do Rio Preto, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Mauro Martins Teixeira
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vivian Vasconcelos Costa
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniele da Glória de Souza
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
42
|
Cottis S, Blisnick AA, Failloux AB, Vernick KD. Determinants of Chikungunya and O'nyong-Nyong Virus Specificity for Infection of Aedes and Anopheles Mosquito Vectors. Viruses 2023; 15:589. [PMID: 36992298 PMCID: PMC10051923 DOI: 10.3390/v15030589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/02/2023] [Accepted: 02/14/2023] [Indexed: 02/23/2023] Open
Abstract
Mosquito-borne diseases caused by viruses and parasites are responsible for more than 700 million infections each year. Anopheles and Aedes are the two major vectors for, respectively, malaria and arboviruses. Anopheles mosquitoes are the primary vector of just one known arbovirus, the alphavirus o'nyong-nyong virus (ONNV), which is closely related to the chikungunya virus (CHIKV), vectored by Aedes mosquitoes. However, Anopheles harbor a complex natural virome of RNA viruses, and a number of pathogenic arboviruses have been isolated from Anopheles mosquitoes in nature. CHIKV and ONNV are in the same antigenic group, the Semliki Forest virus complex, are difficult to distinguish via immunodiagnostic assay, and symptomatically cause essentially the same human disease. The major difference between the arboviruses appears to be their differential use of mosquito vectors. The mechanisms governing this vector specificity are poorly understood. Here, we summarize intrinsic and extrinsic factors that could be associated with vector specificity by these viruses. We highlight the complexity and multifactorial aspect of vectorial specificity of the two alphaviruses, and evaluate the level of risk of vector shift by ONNV or CHIKV.
Collapse
Affiliation(s)
- Solène Cottis
- Genetics and Genomics of Insect Vectors Unit, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris Cité, CNRS UMR2000, F-75015 Paris, France
- Graduate School of Life Sciences ED515, Sorbonne Université UPMC Paris VI, 75252 Paris, France
| | - Adrien A. Blisnick
- Arboviruses and Insect Vectors Unit, Department of Virology, Institut Pasteur, Université de Paris Cité, F-75015 Paris, France
| | - Anna-Bella Failloux
- Arboviruses and Insect Vectors Unit, Department of Virology, Institut Pasteur, Université de Paris Cité, F-75015 Paris, France
| | - Kenneth D. Vernick
- Genetics and Genomics of Insect Vectors Unit, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris Cité, CNRS UMR2000, F-75015 Paris, France
- Graduate School of Life Sciences ED515, Sorbonne Université UPMC Paris VI, 75252 Paris, France
| |
Collapse
|
43
|
Sofyantoro F, Frediansyah A, Priyono DS, Putri WA, Septriani NI, Wijayanti N, Ramadaningrum WA, Turkistani SA, Garout M, Aljeldah M, Al Shammari BR, Alwashmi ASS, Alfaraj AH, Alawfi A, Alshengeti A, Aljohani MH, Aldossary S, Rabaan AA. Growth in chikungunya virus-related research in ASEAN and South Asian countries from 1967 to 2022 following disease emergence: a bibliometric and graphical analysis. Global Health 2023; 19:9. [PMID: 36747262 PMCID: PMC9901127 DOI: 10.1186/s12992-023-00906-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/09/2023] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND ASEAN (Association of Southeast Asian Nations) is composed of ten Southeast Asian countries bound by socio-cultural ties that promote regional peace and stability. South Asia, located in the southern subregion of Asia, includes nine countries sharing similarities in geographical and ethno-cultural factors. Chikungunya is one of the most significant problems in Southeast and South Asian countries. Much of the current chikungunya epidemic in Southeast Asia is caused by the emergence of a virus strain that originated in Africa and spread to Southeast Asia. Meanwhile, in South Asia, three confirmed lineages are in circulation. Given the positive correlation between research activity and the improvement of the clinical framework of biomedical research, this article aimed to examine the growth of chikungunya virus-related research in ASEAN and South Asian countries. METHODS The Scopus database was used for this bibliometric analysis. The retrieved publications were subjected to a number of analyses, including those for the most prolific countries, journals, authors, institutions, and articles. Co-occurrence mapping of terms and keywords was used to determine the current state, emerging topics, and future prospects of chikungunya virus-related research. Bibliometrix and VOSviewer were used to analyze the data and visualize the collaboration network mapping. RESULTS The Scopus search engine identified 1280 chikungunya-related documents published by ASEAN and South Asian countries between 1967 and 2022. According to our findings, India was the most productive country in South Asia, and Thailand was the most productive country in Southeast Asia. In the early stages of the study, researchers investigated the vectors and outbreaks of the chikungunya virus. In recent years, the development of antivirus agents has emerged as a prominent topic. CONCLUSIONS Our study is the first to present the growth of chikungunya virus-related research in ASEAN and South Asian countries from 1967 to 2022. In this study, the evaluation of the comprehensive profile of research on chikungunya can serve as a guide for future studies. In addition, a bibliometric analysis may serve as a resource for healthcare policymakers.
Collapse
Affiliation(s)
- Fajar Sofyantoro
- Faculty of Biology, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
- Center for Tropical Biodiversity, Faculty of Biology, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Andri Frediansyah
- PRTPP, National Research and Innovation Agency (BRIN), Yogyakarta, 55861, Indonesia.
| | - Dwi Sendi Priyono
- Faculty of Biology, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
- Center for Tropical Biodiversity, Faculty of Biology, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | | | | | - Nastiti Wijayanti
- Faculty of Biology, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia.
| | | | | | - Mohammed Garout
- Department of Community Medicine and Health Care for Pilgrims, Faculty of Medicine, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Mohammed Aljeldah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Al Batin, Hafr Al Batin, 39831, Saudi Arabia
| | - Basim R Al Shammari
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Al Batin, Hafr Al Batin, 39831, Saudi Arabia
| | - Ameen S S Alwashmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, 51452, Saudi Arabia
| | - Amal H Alfaraj
- Pediatric Department, Abqaiq General Hospital, First Eastern Health Cluster, Abqaiq, 33261, Saudi Arabia
| | - Abdulsalam Alawfi
- Department of Pediatrics, College of Medicine, Taibah University, Al-Madinah, 41491, Saudi Arabia
| | - Amer Alshengeti
- Department of Pediatrics, College of Medicine, Taibah University, Al-Madinah, 41491, Saudi Arabia
- Department of Infection Prevention and Control, Prince Mohammad Bin Abdulaziz Hospital, National Guard Health Affairs, Al-Madinah, 41491, Saudi Arabia
| | - Maha H Aljohani
- Department of infectious diseases, King Fahad Hospital, Madinah, 42351, Saudi Arabia
| | - Sahar Aldossary
- Pediatric Infectious Diseases, Women and Children's Health Institute, Johns Hopkins Aramco Healthcare, Dhahran, 31311, Saudi Arabia
| | - Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, 31311, Saudi Arabia.
- College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia.
- Department of Public Health and Nutrition, The University of Haripur, Haripur, 22610, Pakistan.
| |
Collapse
|
44
|
Koh C, Frangeul L, Blanc H, Ngoagouni C, Boyer S, Dussart P, Grau N, Girod R, Duchemin JB, Saleh MC. Ribosomal RNA (rRNA) sequences from 33 globally distributed mosquito species for improved metagenomics and species identification. eLife 2023; 12:82762. [PMID: 36688360 PMCID: PMC10014081 DOI: 10.7554/elife.82762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Total RNA sequencing (RNA-seq) is an important tool in the study of mosquitoes and the RNA viruses they vector as it allows assessment of both host and viral RNA in specimens. However, there are two main constraints. First, as with many other species, abundant mosquito ribosomal RNA (rRNA) serves as the predominant template from which sequences are generated, meaning that the desired host and viral templates are sequenced far less. Second, mosquito specimens captured in the field must be correctly identified, in some cases to the sub-species level. Here, we generate mosquito rRNA datasets which will substantially mitigate both of these problems. We describe a strategy to assemble novel rRNA sequences from mosquito specimens and produce an unprecedented dataset of 234 full-length 28S and 18S rRNA sequences of 33 medically important species from countries with known histories of mosquito-borne virus circulation (Cambodia, the Central African Republic, Madagascar, and French Guiana). These sequences will allow both physical and computational removal of rRNA from specimens during RNA-seq protocols. We also assess the utility of rRNA sequences for molecular taxonomy and compare phylogenies constructed using rRNA sequences versus those created using the gold standard for molecular species identification of specimens-the mitochondrial cytochrome c oxidase I (COI) gene. We find that rRNA- and COI-derived phylogenetic trees are incongruent and that 28S and concatenated 28S+18S rRNA phylogenies reflect evolutionary relationships that are more aligned with contemporary mosquito systematics. This significant expansion to the current rRNA reference library for mosquitoes will improve mosquito RNA-seq metagenomics by permitting the optimization of species-specific rRNA depletion protocols for a broader range of species and streamlining species identification by rRNA sequence and phylogenetics.
Collapse
Affiliation(s)
- Cassandra Koh
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Viruses and RNA Interference Unit, F-75015ParisFrance
| | - Lionel Frangeul
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Viruses and RNA Interference Unit, F-75015ParisFrance
| | - Hervé Blanc
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Viruses and RNA Interference Unit, F-75015ParisFrance
| | - Carine Ngoagouni
- Institut Pasteur de Bangui, Medical Entomology LaboratoryBanguiCentral African Republic
| | - Sébastien Boyer
- Institut Pasteur du Cambodge, Medical and Veterinary Entomology UnitPhnom PenhCambodia
| | | | - Nina Grau
- Institut Pasteur de Madagascar, Medical Entomology UnitAntananarivoMadagascar
| | - Romain Girod
- Institut Pasteur de Madagascar, Medical Entomology UnitAntananarivoMadagascar
| | - Jean-Bernard Duchemin
- Institut Pasteur de la Guyane, Vectopôle Amazonien Emile AbonnencCayenneFrench Guiana
| | - Maria-Carla Saleh
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Viruses and RNA Interference Unit, F-75015ParisFrance
| |
Collapse
|
45
|
Potential Mammalian Vector-Borne Diseases in Live and Wet Markets in Indonesia and Myanmar. MICROBIOLOGY RESEARCH 2023. [DOI: 10.3390/microbiolres14010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Vector-borne diseases spread from wild animals and their associated ectoparasites to humans and domesticated animals. Wildlife markets are recognized as important areas where this transfer can take place. We assessed the potential for spreading vector-borne diseases in two live and wet markets in Myanmar (Mong La, on the Myanmar-China border) and Indonesia (Sukahaji in Bandung on the island of Java) by making an inventory of all live and freshly killed wild mammals for sale. For eight mammal families, we quantified the number of animals on offer, and we used a heatmap cluster analysis to map vector-borne diseases that these families may carry. In Myanmar, we observed large numbers of wild pigs and deer (potentially carrying West Nile and various encephalitis viruses) whereas in Indonesia we observed Old World fruit bats (potentially carrying Chikungunya and encephalitis viruses) and squirrels (potentially carrying West Nile and encephalitis viruses). The trade in Indonesia was dominated by live mammals offered for sale as pets, and only Old World fruit bats and squirrels traded for traditional Asian medicine were killed in the markets. The trade in Myanmar was more geared towards wild meat (e.g., wild pigs, deer, primates) and traditional Asian medicine (squirrels). The combined risks of vector-borne diseases spreading from traded animals to human health highlight the need for an integrated approach protecting public health, economic interests and biodiversity.
Collapse
|
46
|
Hakim MS, Aman AT. Understanding the Biology and Immune Pathogenesis of Chikungunya Virus Infection for Diagnostic and Vaccine Development. Viruses 2022; 15:48. [PMID: 36680088 PMCID: PMC9863735 DOI: 10.3390/v15010048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Chikungunya virus, the causative agent of chikungunya fever, is generally characterized by the sudden onset of symptoms, including fever, rash, myalgia, and headache. In some patients, acute chikungunya virus infection progresses to severe and chronic arthralgia that persists for years. Chikungunya infection is more commonly identified in tropical and subtropical regions. However, recent expansions and epidemics in the temperate regions have raised concerns about the future public health impact of chikungunya diseases. Several underlying factors have likely contributed to the recent re-emergence of chikungunya infection, including urbanization, human travel, viral adaptation to mosquito vectors, lack of effective control measures, and the spread of mosquito vectors to new regions. However, the true burden of chikungunya disease is most likely to be underestimated, particularly in developing countries, due to the lack of standard diagnostic assays and clinical manifestations overlapping with those of other endemic viral infections in the regions. Additionally, there have been no chikungunya vaccines available to prevent the infection. Thus, it is important to update our understanding of the immunopathogenesis of chikungunya infection, its clinical manifestations, the diagnosis, and the development of chikungunya vaccines.
Collapse
Affiliation(s)
- Mohamad S. Hakim
- Department of Microbiology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | | |
Collapse
|
47
|
Millsapps EM, Underwood EC, Barr KL. Development and Application of Treatment for Chikungunya Fever. Res Rep Trop Med 2022; 13:55-66. [PMID: 36561535 PMCID: PMC9767026 DOI: 10.2147/rrtm.s370046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
The development and application of treatment for Chikungunya fever (CHIKF) remains complicated as there is no current standard treatment and many barriers to research exist. Chikungunya virus (CHIKV) causes serious global health implications due to its socioeconomic impact and high morbidity rates. In research, treatment through natural and pharmaceutical techniques is being evaluated for their efficacy and effectiveness. Natural treatment options, such as homeopathy and physiotherapy, give patients a variety of options for how to best manage acute and chronic symptoms. Some of the most used pharmaceutical therapies for CHIKV include non-steroidal anti-inflammatory drugs (NSAIDS), methotrexate (MTX), chloroquine, and ribavirin. Currently, there is no commercially available vaccine for chikungunya, but vaccine development is crucial for this virus. Potential treatments need further research until they can become a standard part of treatment. The barriers to research for this complicated virus create challenges in the efficacy and equitability of its research. The rising need for increased research to fully understand chikungunya in order to develop more effective treatment options is vital in protecting endemic populations globally.
Collapse
Affiliation(s)
- Erin M Millsapps
- Center for Global Health and Infectious Disease Research, University of South Florida, Tampa, FL, USA
| | - Emma C Underwood
- Center for Global Health and Infectious Disease Research, University of South Florida, Tampa, FL, USA
| | - Kelli L Barr
- Center for Global Health and Infectious Disease Research, University of South Florida, Tampa, FL, USA,Correspondence: Kelli L Barr, Center for Global Health and Infectious Disease Research, University of South Florida, 3720 Spectrum Blvd. Suite 304, Tampa, FL, 33612, USA, Tel +1 813 974 4480, Fax +1 813 974 4962, Email
| |
Collapse
|
48
|
Hakim MS, Annisa L, Gazali FM, Aman AT. The origin and continuing adaptive evolution of chikungunya virus. Arch Virol 2022; 167:2443-2455. [PMID: 35987965 DOI: 10.1007/s00705-022-05570-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/05/2022] [Indexed: 12/14/2022]
Abstract
Chikungunya virus (CHIKV) is the responsible agent of chikungunya fever, a debilitating arthritic disease in humans. CHIKV is endemic in Africa and Asia, although transmission cycles are considerably different on these continents. Before 2004, CHIKV had received little attention, since it was only known to cause localised outbreaks in a limited region with no fatalities. However, the recent global reemergence of CHIKV has caused serious global health problems and shown its potential to become a significant viral threat in the future. Unexpectedly, the reemergence is more rapid and is geographically more extensive, especially due to increased intensity of global travel systems or failure to contain mosquito populations. Another important factor is the successful adaptation of CHIKV to a new vector, the Aedes albopictus mosquito. Ae. albopictus survives in both temperate and tropical climates, thus facilitating CHIKV expansion to non-endemic regions. The continuous spread and transmission of CHIKV pose challenges for the development of effective vaccines and specific antiviral therapies. In this review, we discuss the biology and origin of CHIKV in Africa as well as its subsequent expansion to other parts of the world. We also review the transmission cycle of CHIKV and its continuing adaptation to its mosquito vectors and vertebrate hosts. More-complete understanding of the continuous evolution of CHIKV may help in predicting the emergence of CHIKV strains with possibly greater transmission efficiency in the future.
Collapse
Affiliation(s)
- Mohamad S Hakim
- Department of Microbiology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia.
| | - Luthvia Annisa
- Department of Microbiology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Faris M Gazali
- Master Program in Biotechnology, Postgraduate School, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Abu T Aman
- Department of Microbiology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| |
Collapse
|
49
|
Babaeimarzangou SS, Zaker H, Soleimannezhadbari E, Gamchi NS, Kazeminia M, Tarighi S, Seyedian H, Tsatsakis A, Spandidos DA, Margina D. Vaccine development for zoonotic viral diseases caused by positive‑sense single‑stranded RNA viruses belonging to the Coronaviridae and Togaviridae families (Review). Exp Ther Med 2022; 25:42. [PMID: 36569444 PMCID: PMC9768462 DOI: 10.3892/etm.2022.11741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022] Open
Abstract
Outbreaks of zoonotic viral diseases pose a severe threat to public health and economies worldwide, with this currently being more prominent than it previously was human history. These emergency zoonotic diseases that originated and transmitted from vertebrates to humans have been estimated to account for approximately one billion cases of illness and have caused millions of deaths worldwide annually. The recent emergence of severe acute respiratory syndrome coronavirus-2 (coronavirus disease 2019) is an excellent example of the unpredictable public health threat causing a pandemic. The present review summarizes the literature data regarding the main vaccine developments in human clinical phase I, II and III trials against the zoonotic positive-sense single-stranded RNA viruses belonging to the Coronavirus and Alphavirus genera, including severe acute respiratory syndrome, Middle east respiratory syndrome, Venezuelan equine encephalitis virus, Semliki Forest virus, Ross River virus, Chikungunya virus and O'nyong-nyong virus. That there are neither vaccines nor effective antiviral drugs available against most of these viruses is undeniable. Therefore, new explosive outbreaks of these zoonotic viruses may surely be expected. The present comprehensive review provides an update on the status of vaccine development in different clinical trials against these viruses, as well as an overview of the present results of these trials.
Collapse
Affiliation(s)
- Seyed Sajjad Babaeimarzangou
- Division of Poultry Health and Diseases, Department of Clinical Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia 5756151818, Iran
| | - Himasadat Zaker
- Histology and Microscopic Analysis Division, RASTA Specialized Research Institute (RSRI), West Azerbaijan Science and Technology Park (WASTP), Urmia 5756115322, Iran
| | | | - Naeimeh Shamsi Gamchi
- Histology and Microscopic Analysis Division, RASTA Specialized Research Institute (RSRI), West Azerbaijan Science and Technology Park (WASTP), Urmia 5756115322, Iran
| | - Masoud Kazeminia
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417935840, Iran
| | - Shima Tarighi
- Veterinary Office of West Azerbaijan Province, Urmia 5717617695, Iran
| | - Homayon Seyedian
- Faculty of Veterinary Medicine, Urmia University, Urmia 5756151818, Iran
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, Department of Medicine, University of Crete, 71307 Heraklion, Greece,Correspondence to: Professor Denisa Margina, Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Denisa Margina
- Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 020956 Bucharest, Romania,Correspondence to: Professor Denisa Margina, Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| |
Collapse
|
50
|
Genomic Epidemiology Reveals the Circulation of the Chikungunya Virus East/Central/South African Lineage in Tocantins State, North Brazil. Viruses 2022; 14:v14102311. [PMID: 36298867 PMCID: PMC9611869 DOI: 10.3390/v14102311] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
The chikungunya virus (CHIKV) is a mosquito-borne virus of the family Togaviridae transmitted to humans by Aedes spp. mosquitoes. In Brazil, imported cases have been reported since June 2014 through two independent introductions, one caused by Asian Lineage in Oiapoque, Amapá state, North Region, and another caused by East/Central/South African (ECSA) in Feira de Santana, Bahia state, Northeast Region. Moreover, there is still limited information about the genomic epidemiology of the CHIKV from surveillance studies. The Tocantins state, located in Northern Brazil, reported an increase in the number of CHIKV cases at the end of 2021 and the beginning of 2022. Thus, to better understand the dispersion dynamics of this viral pathogen in the state, we generated 27 near-complete CHIKV genome sequences from four cities, obtained from clinical samples. Our results showed that the newly CHIKV genomes from Tocantins belonged to the ECSA lineage. Phylogenetic reconstruction revealed that Tocantins' strains formed a single well-supported clade, which appear to be closely related to isolates from the Rio Grande do Norte state (Northeast Brazil) and the Rio de Janeiro state (Southeast Brazil), that experienced an explosive ECSA epidemic between 2016-2019. Mutation analyses showed eleven frequent non-synonymous mutations in the structural and non-structural proteins, indicating the autochthonous transmission of the CHIKV in the state. None of the genomes recovered within the Tocantins samples carry the A226V mutation in the E1 protein associated with increased transmission in A. albopictus. The study presented here highlights the importance of continued genomic surveillance to provide information not only on recording mutations along the viral genome but as a molecular surveillance tool to trace virus spread within the country, to predict events of likely occurrence of new infections, and, as such, contribute to an improved public health service.
Collapse
|