1
|
Andreolla AP, Koishi AC, Borges AA, de Oliveira LA, de Oliveira VG, Lima NM, Ávila EP, de Castro PP, Amarante GW, de Almeida MV, Bordignon J, Duarte dos Santos CN. Identification and Characterization of Antiviral Activity of Synthetic Compounds Against Mayaro Virus. Pharmaceuticals (Basel) 2025; 18:717. [PMID: 40430536 PMCID: PMC12115251 DOI: 10.3390/ph18050717] [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: 04/02/2025] [Revised: 05/04/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Background/objectives: In Brazil, the co-circulation of arboviruses-such as dengue, Zika, yellow fever, and Chikungunya viruses-creates a complex epidemiological landscape, drawing attention from health authorities due to high morbidity and mortality rates. Also present in this context is the Mayaro virus (MAYV), a neglected arbovirus, which can also cause severe syndromes and has been expanding beyond its usual endemic areas in northern and central-western Brazil. Epidemiological surveillance measures remain limited, and there are no effective prophylactic strategies or antiviral treatments for this neglected arbovirus. In this study, we evaluated the antiviral activity of commercial and synthetic compounds against MAYV using an image high-throughput screening (iHTS) system. Methods: A total of 52 compounds from an FDA-approved commercial library (Tocriscreen) and 50 other compounds were tested. Results: Seven compounds showed anti-MAYV activity and were non-toxic for the following cell lines: Naringenin, LLA9A, chrysin, and its ester C6. Post-infection treatments with these selected compounds significantly decreased the percentage of infected cells and the release of infectious viral particles in the supernatant. Additionally, anti-MAYV activity of these four selected hits was confirmed using several human cell lines and two different MAYV genotypes. Conclusions: Our results indicate that the iHTS platform is effective for screening anti-MAYV drugs and that four promising compounds can efficiently inhibit MAYV replication in human cell lines. Although in vivo studies are still required to confirm the efficacy of the selected hits, our findings provide a starting point for developing a potential treatment for MAYV infections.
Collapse
Affiliation(s)
- Ana Paula Andreolla
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, ICC/Fiocruz, Rua Prof. Algacyr Munhoz Mader 3775, Cidade Industrial de Curitiba, Curitiba 81350-010, Paraná, Brazil; (A.P.A.); (A.C.K.)
| | - Andrea Cristine Koishi
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, ICC/Fiocruz, Rua Prof. Algacyr Munhoz Mader 3775, Cidade Industrial de Curitiba, Curitiba 81350-010, Paraná, Brazil; (A.P.A.); (A.C.K.)
| | - Alessandra Abel Borges
- Laboratório de Pesquisas em Virologia e Imunologia, Instituto de Ciências Biológicas e da Saúde (ICBS), Universidade Federal de Alagoas (UFAL), Av. Lourival Melo Mota, s/n, Tabuleiro do Martins, Maceió 57072-900, Alagoas, Brazil;
| | - Larissa Albuquerque de Oliveira
- Departamento de Química, ICE, Universidade Federal de Juiz de Fora, Rua José Lourenço Kelmer s/n, Martelos, Juiz de Fora 36036-900, Minas Gerais, Brazil; (L.A.d.O.); (N.M.L.); (E.P.Á.); (P.P.d.C.); (G.W.A.); (M.V.d.A.)
| | - Viviane Guedes de Oliveira
- Instituto de Educação, Agricultura e Ambiente, IEAA, Universidade Federal do Amazonas, Rua 29 de agosto, Centro, Humaitá 69800–000, Amazonas, Brazil;
| | - Nerilson Marques Lima
- Departamento de Química, ICE, Universidade Federal de Juiz de Fora, Rua José Lourenço Kelmer s/n, Martelos, Juiz de Fora 36036-900, Minas Gerais, Brazil; (L.A.d.O.); (N.M.L.); (E.P.Á.); (P.P.d.C.); (G.W.A.); (M.V.d.A.)
| | - Eloah Pereira Ávila
- Departamento de Química, ICE, Universidade Federal de Juiz de Fora, Rua José Lourenço Kelmer s/n, Martelos, Juiz de Fora 36036-900, Minas Gerais, Brazil; (L.A.d.O.); (N.M.L.); (E.P.Á.); (P.P.d.C.); (G.W.A.); (M.V.d.A.)
| | - Pedro Pôssa de Castro
- Departamento de Química, ICE, Universidade Federal de Juiz de Fora, Rua José Lourenço Kelmer s/n, Martelos, Juiz de Fora 36036-900, Minas Gerais, Brazil; (L.A.d.O.); (N.M.L.); (E.P.Á.); (P.P.d.C.); (G.W.A.); (M.V.d.A.)
| | - Giovanni Wilson Amarante
- Departamento de Química, ICE, Universidade Federal de Juiz de Fora, Rua José Lourenço Kelmer s/n, Martelos, Juiz de Fora 36036-900, Minas Gerais, Brazil; (L.A.d.O.); (N.M.L.); (E.P.Á.); (P.P.d.C.); (G.W.A.); (M.V.d.A.)
| | - Mauro Vieira de Almeida
- Departamento de Química, ICE, Universidade Federal de Juiz de Fora, Rua José Lourenço Kelmer s/n, Martelos, Juiz de Fora 36036-900, Minas Gerais, Brazil; (L.A.d.O.); (N.M.L.); (E.P.Á.); (P.P.d.C.); (G.W.A.); (M.V.d.A.)
| | - Juliano Bordignon
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, ICC/Fiocruz, Rua Prof. Algacyr Munhoz Mader 3775, Cidade Industrial de Curitiba, Curitiba 81350-010, Paraná, Brazil; (A.P.A.); (A.C.K.)
| | - Claudia Nunes Duarte dos Santos
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, ICC/Fiocruz, Rua Prof. Algacyr Munhoz Mader 3775, Cidade Industrial de Curitiba, Curitiba 81350-010, Paraná, Brazil; (A.P.A.); (A.C.K.)
| |
Collapse
|
2
|
Lantz AM, Baxter VK. Neuropathogenesis of Old World Alphaviruses: Considerations for the Development of Medical Countermeasures. Viruses 2025; 17:261. [PMID: 40007016 PMCID: PMC11860675 DOI: 10.3390/v17020261] [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: 01/17/2025] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Chikungunya virus (CHIKV) and other alphaviruses that primarily induce arthritogenic disease in humans, known as "Old World" alphaviruses, present an emerging public health concern as geographic ranges of mosquito vectors expand due to climate change. While a vaccine against CHIKV has recently been approved by several countries in North America and Europe, access to effective preventative countermeasures against disease induced by Old World alphaviruses remains elusive for the most vulnerable populations. Furthermore, treatment options continue to be limited to supportive care. Atypical neurological disease manifestations caused by Old World alphaviruses, which make up as many as 25% of the cases in some CHIKV outbreaks, present special challenges when considering strategies for developing effective countermeasures. This review focuses on Old World alphaviruses, specifically CHIKV, Ross River virus, O'nyoug-nyoug virus, and Mayaro virus, concentrating on the atypical neurological disease manifestations they may cause. Our current understanding of Old World alphavirus neuropathogenesis, gained from human cases and preclinical animal models, is discussed, including viral and host factors' roles in disease development. The current state of alphavirus preventatives and treatments, both virus-targeting and host-directed therapies, is then summarized and discussed in the context of addressing neurological disease induced by Old World alphaviruses.
Collapse
|
3
|
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
|
4
|
Porier DL, Adam A, Kang L, Michalak P, Tupik J, Santos MA, Tanelus M, López K, Auguste DI, Lee C, Allen IC, Wang T, Auguste AJ. Humoral and T-cell-mediated responses to an insect-specific flavivirus-based Zika virus vaccine candidate. PLoS Pathog 2024; 20:e1012566. [PMID: 39388457 PMCID: PMC11495591 DOI: 10.1371/journal.ppat.1012566] [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: 12/13/2023] [Revised: 10/22/2024] [Accepted: 09/05/2024] [Indexed: 10/12/2024] Open
Abstract
Flaviviruses represent a significant global health threat and relatively few licensed vaccines exist to protect against them. Insect-specific flaviviruses (ISFVs) are incapable of replication in humans and have emerged as a novel and promising tool for flavivirus vaccine development. ISFV-based flavivirus vaccines have shown exceptional safety, immunogenicity, and efficacy, however, a detailed assessment of the correlates of protection and immune responses induced by these vaccines are still needed for vaccine optimization. Here, we explore the mechanisms of protective immunity induced by a previously created pre-clinical Zika virus (ZIKV) vaccine candidate, called Aripo/Zika (ARPV/ZIKV). In brief, immunocompromised IFN-αβR-/- mice passively immunized with ARPV/ZIKV immune sera experienced protection after lethal ZIKV challenge, although this protection was incomplete. ARPV/ZIKV-vaccinated IFN-αβR-/- mice depleted of CD4+ or CD8+ T-cells at the time of ZIKV challenge showed no morbidity or mortality. However, the adoptive transfer of ARPV/ZIKV-primed T-cells into recipient IFN-αβR-/- mice resulted in a two-day median increase in survival time compared to controls. Altogether, these results suggest that ARPV/ZIKV-induced protection is primarily mediated by neutralizing antibodies at the time of challenge and that T-cells may play a comparatively minor but cumulative role in the protection observed. Lastly, ARPV/ZIKV-vaccinated Tcra KO mice, which are deficient in T-cell responses, experienced significant mortality post-challenge. These results suggest that ARPV/ZIKV-induced cell-mediated responses are critical for development of protective immune responses at vaccination. Despite the strong focus on neutralizing antibody responses to novel flavivirus vaccine candidates, these results suggest that cell-mediated responses induced by ISFV-based vaccines remain important to overall protective responses.
Collapse
Affiliation(s)
- Danielle L. Porier
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Awadalkareem Adam
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Lin Kang
- Department of Biomedical Research, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States of America
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
- College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, United States of America
| | - Pawel Michalak
- Department of Biomedical Research, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States of America
- Center for One Health Research, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
- Institute of Evolution, University of Haifa, Haifa, Israel
| | - Juselyn Tupik
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
| | - Matthew A. Santos
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Manette Tanelus
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Krisangel López
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Dawn I. Auguste
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Christy Lee
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Irving C. Allen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Albert J. Auguste
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
| |
Collapse
|
5
|
Weber WC, Andoh TF, Kreklywich CN, Streblow ZJ, Denton M, Streblow MM, Powers JM, Sulgey G, Medica S, Dmitriev I, Curiel DT, Haese NN, Streblow DN. Nonreciprocity in CHIKV and MAYV Vaccine-Elicited Protection. Vaccines (Basel) 2024; 12:970. [PMID: 39340002 PMCID: PMC11435824 DOI: 10.3390/vaccines12090970] [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: 07/19/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Chikungunya virus (CHIKV) is a pathogenic arthritogenic alphavirus responsible for large-scale human epidemics for which a vaccine was recently approved for use. Mayaro virus (MAYV) is a related emerging alphavirus with epidemic potential with circulation overlap potential with CHIKV. We previously reported the ability of a non-replicating human adenovirus (AdV)-vectored vaccine expressing the MAYV structural polyprotein to protect against disease in mice following challenge with MAYV, CHIKV and UNAV. Herein, we evaluated mouse immunity and protective efficacy for an AdV-CHIKV full structural polyprotein vaccine in combination with heterologous AdV-MAYV prime/boost regimens versus vaccine coadministration. Heterologous prime/boost regimens skewed immunity toward the prime vaccine antigen but allowed for a boost of cross-neutralizing antibodies, while vaccine co-administration elicited robust, balanced responses capable of boosting. All immunization strategies protected against disease from homologous virus infection, but reciprocal protective immunity differences were revealed upon challenge with heterologous viruses. In vivo passive transfer experiments reproduced the inequity in reciprocal cross-protection after heterologous MAYV challenge. We detected in vitro antibody-dependent enhancement of MAYV replication, suggesting a potential mechanism for the lack of cross-protection. Our findings provide important insights into rational alphavirus vaccine design that may have important implications for the evolving alphavirus vaccine landscape.
Collapse
Affiliation(s)
- Whitney C. Weber
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Takeshi F. Andoh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - Craig N. Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - Zachary J. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - Michael Denton
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - Magdalene M. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - John M. Powers
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - Gauthami Sulgey
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - Samuel Medica
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - Igor Dmitriev
- Cancer Biology Division, Department of Radiation Oncology, Washington University, St. Louis, MO 63110, USA; (I.D.); (D.T.C.)
| | - David T. Curiel
- Cancer Biology Division, Department of Radiation Oncology, Washington University, St. Louis, MO 63110, USA; (I.D.); (D.T.C.)
| | - Nicole N. Haese
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - Daniel N. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| |
Collapse
|
6
|
Wei LLL, Tom R, Kim YC. Mayaro Virus: An Emerging Alphavirus in the Americas. Viruses 2024; 16:1297. [PMID: 39205271 PMCID: PMC11359717 DOI: 10.3390/v16081297] [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: 04/29/2024] [Revised: 07/25/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Mayaro virus (MAYV) is an arbovirus first isolated in Trinidad and Tobago in 1954. MAYV is the causative agent of Mayaro fever, which is characterised by high fever, maculopapular rash, myalgia and arthralgia. The potential for chronic arthralgia is of particular clinical concern. Currently, MAYV outbreaks are restricted to South and Central America, with some cases reported in Africa as well as several imported cases in Europe. However, in recent years, MAYV has become a growing global concern due to its potential to emerge into urban transmission cycles. Challenges faced with diagnostics, as well as a lack of specific antivirals or licensed vaccines further exacerbate the potential global health threat posed by MAYV. In this review, we discuss this emerging arboviral threat with a particular focus on the current treatment and vaccine development efforts. Overall, MAYV remains a neglected arbovirus due to its limited area of transmission. However, with the potential of its urbanisation and expanding circulation, the threat MAYV poses to global health cannot be overlooked. Further research into the improvement of current diagnostics, as well as the development of efficacious antivirals and vaccines will be crucial to help prevent and manage potential MAYV outbreaks.
Collapse
Affiliation(s)
- Lily Li Lin Wei
- Somerville College, University of Oxford, Woodstock Road, Oxford OX2 6HD, UK; (L.L.L.W.); (R.T.)
| | - Rufaro Tom
- Somerville College, University of Oxford, Woodstock Road, Oxford OX2 6HD, UK; (L.L.L.W.); (R.T.)
| | - Young Chan Kim
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford OX3 7LE, UK
- Centre for Human Genetics, Division of Structural Biology, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| |
Collapse
|
7
|
Hameed M, Solomon NA, Weger-Lucarelli J. Lack of pathogenic involvement of CCL4 and its receptor CCR5 in arthritogenic alphavirus disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.606106. [PMID: 39131287 PMCID: PMC11312581 DOI: 10.1101/2024.07.31.606106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Arthritogenic alphaviruses, including chikungunya virus (CHIKV), Mayaro virus (MAYV), Ross River virus (RRV), and O'nyong nyong virus (ONNV) are emerging and reemerging viruses that cause disease characterized by fever, rash, and incapacitating joint swelling. Alphavirus infection induces robust immune responses in infected hosts, leading to the upregulation of several cytokines and chemokines, including chemokine C ligand 4 (CCL4). CCL4 is a chemoattractant for immune cells such as T cells, natural killer cells, monocytes/macrophages, and dendritic cells, recruiting these cells to the site of infection, stimulating the release of proinflammatory mediators, and inducing T cell differentiation. CCL4 has been found at high levels in both the acute and chronic phases of chikungunya disease; however, the role of CCL4 in arthritogenic alphavirus disease development remains unexplored. Here, we tested the effect of CCL4 on MAYV infection in mice through antibody depletion and treatment with recombinant mouse CCL4. We observed no differences in mice depleted of CCL4 or treated with recombinant CCL4 in terms of disease progression such as weight loss and footpad swelling or the development of viremia. CCL4 uses the G protein-coupled receptor C-C chemokine receptor type 5 (CCR5). To determine whether CCR5 deficiency would alter disease outcomes or virus replication in mice, we inoculated CCR5 knockout (CCR5-/-) mice with MAYV and observed no effect on disease development and immune cell profile of blood and footpads between CCR5-/- and wild type mice. These studies failed to identify a clear role for CCL4 or its receptor CCR5 in MAYV infection.
Collapse
Affiliation(s)
- Muddassar Hameed
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - Norman A. Solomon
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
- Lead contact
| |
Collapse
|
8
|
Hamilton MM, Webb EM, Peterson MC, Patel G, Porto M, Orekov T, Erasmus JH, Finneyfrock B, Cook A, Auguste AJ, Kar S. Comparative pathogenesis of three Mayaro virus genotypes in the cynomolgus macaque. J Gen Virol 2024; 105. [PMID: 38995674 DOI: 10.1099/jgv.0.002001] [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] [Indexed: 07/13/2024] Open
Abstract
Mayaro virus (MAYV), a mosquito-borne alphavirus, is considered an emerging threat to public health with epidemic potential. Phylogenetic studies show the existence of three MAYV genotypes. In this study, we provide a preliminary analysis of the pathogenesis of all three MAYV genotypes in cynomolgus macaques (Macaca facicularis, Mauritian origin). Significant MAYV-specific RNAemia and viremia were detected during acute infection in animals challenged intravenously with the three MAYV genotypes, and strong neutralizing antibody responses were observed. MAYV RNA was detected at high levels in lymphoid tissues, joint muscle and synovia over 1 month after infection, suggesting that this model could serve as a promising tool in studying MAYV-induced chronic arthralgia, which can persist for years. Significant leucopenia was observed across all MAYV genotypes, peaking with RNAemia. Notable differences in the severity of acute RNAemia and composition of cytokine responses were observed among the three MAYV genotypes. Our model showed no outward signs of clinical disease, but several major endpoints for future MAYV pathology and intervention studies are described. Disruptions to normal blood cell counts and cytokine responses were markedly distinct from those observed in macaque models of CHIKV infection, underlining the importance of developing non-human primate models specific to MAYV infection.
Collapse
Affiliation(s)
| | - Emily M Webb
- Department of Entomology, College of Agriculture and Life Sciences, Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | | | | | | | | | | | | | | | - Albert J Auguste
- Department of Entomology, College of Agriculture and Life Sciences, Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | | |
Collapse
|
9
|
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
|
10
|
Weber WC, Labriola CS, Kreklywich CN, Ray K, Haese NN, Andoh TF, Denton M, Medica S, Streblow MM, Smith PP, Mizuno N, Frias N, Fisher MB, Barber-Axthelm AM, Chun K, Uttke S, Whitcomb D, DeFilippis V, Rakshe S, Fei SS, Axthelm MK, Smedley JV, Streblow DN. Mayaro virus pathogenesis and immunity in rhesus macaques. PLoS Negl Trop Dis 2023; 17:e0011742. [PMID: 37983245 PMCID: PMC10695392 DOI: 10.1371/journal.pntd.0011742] [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: 06/06/2023] [Revised: 12/04/2023] [Accepted: 10/19/2023] [Indexed: 11/22/2023] Open
Abstract
Mayaro virus (MAYV) is a mosquito-transmitted alphavirus that causes debilitating and persistent arthritogenic disease. While MAYV was previously reported to infect non-human primates (NHP), characterization of MAYV pathogenesis is currently lacking. Therefore, in this study we characterized MAYV infection and immunity in rhesus macaques. To inform the selection of a viral strain for NHP experiments, we evaluated five MAYV strains in C57BL/6 mice and showed that MAYV strain BeAr505411 induced robust tissue dissemination and disease. Three male rhesus macaques were subcutaneously challenged with 105 plaque-forming units of this strain into the arms. Peak plasma viremia occurred at 2 days post-infection (dpi). NHPs were taken to necropsy at 10 dpi to assess viral dissemination, which included the muscles and joints, lymphoid tissues, major organs, male reproductive tissues, as well as peripheral and central nervous system tissues. Histological examination demonstrated that MAYV infection was associated with appendicular joint and muscle inflammation as well as presence of perivascular inflammation in a wide variety of tissues. One animal developed a maculopapular rash and two NHP had viral RNA detected in upper torso skin samples, which was associated with the presence of perivascular and perifollicular lymphocytic aggregation. Analysis of longitudinal peripheral blood samples indicated a robust innate and adaptive immune activation, including the presence of anti-MAYV neutralizing antibodies with activity against related Una virus and chikungunya virus. Inflammatory cytokines and monocyte activation also peaked coincident with viremia, which was well supported by our transcriptomic analysis highlighting enrichment of interferon signaling and other antiviral processes at 2 days post MAYV infection. The rhesus macaque model of MAYV infection recapitulates many of the aspects of human infection and is poised to facilitate the evaluation of novel therapies and vaccines targeting this re-emerging virus.
Collapse
Affiliation(s)
- Whitney C. Weber
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Caralyn S. Labriola
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Craig N. Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Karina Ray
- Bioinformatics & Biostatistics Core, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Nicole N. Haese
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Takeshi F. Andoh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Michael Denton
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Samuel Medica
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Magdalene M. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Patricia P. Smith
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Nobuyo Mizuno
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Nina Frias
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Miranda B. Fisher
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Aaron M. Barber-Axthelm
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Kimberly Chun
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Samantha Uttke
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Danika Whitcomb
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Victor DeFilippis
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Shauna Rakshe
- Bioinformatics & Biostatistics Core, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Suzanne S. Fei
- Bioinformatics & Biostatistics Core, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Michael K. Axthelm
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Jeremy V. Smedley
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Daniel N. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| |
Collapse
|
11
|
Rosa RB, de Castro EF, de Oliveira Santos D, da Silva MV, Pena LJ. Mouse Models of Mayaro Virus. Viruses 2023; 15:1803. [PMID: 37766210 PMCID: PMC10534528 DOI: 10.3390/v15091803] [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: 05/30/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 09/29/2023] Open
Abstract
Mayaro virus (MAYV), the etiologic agent of Mayaro fever, leads patients to severe myalgia and arthralgia, which can have a major impact on public health in all the countries where the virus circulates. The emergence and dissemination of new viruses have led the scientific community to develop new in vivo models that can help in the fight against new diseases. So far, mice have been the most used animal model in studies with MAYV and have proved to be an adequate model for recapitulating several aspects of the disease observed in humans. Mice are widely used in in vivo research and, therefore, are well known in the scientific community, which has allowed for different strains to be investigated in the study of MAYV. In this review, we summarize the main studies with MAYV using mice as an experimental model and discuss how they can contribute to the advancement of the understanding of its pathogenesis and the development of new drugs and vaccines.
Collapse
Affiliation(s)
- Rafael Borges Rosa
- Department of Virology and Experimental Therapy (LAVITE), Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife 50740-465, Brazil
- Rodents Animal Facilities Complex, Federal University of Uberlandia (REBIR-UFU), Uberlandia 38400-902, Brazil;
| | - Emilene Ferreira de Castro
- Faculty of Medicine, Federal University of Uberlandia, Uberlandia 38400-902, Brazil;
- Dental Hospital, Oral Pathology Laboratory, University of Uberlandia, Uberlandia 38400-902, Brazil;
| | | | - Murilo Vieira da Silva
- Rodents Animal Facilities Complex, Federal University of Uberlandia (REBIR-UFU), Uberlandia 38400-902, Brazil;
| | - Lindomar José Pena
- Department of Virology and Experimental Therapy (LAVITE), Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife 50740-465, Brazil
| |
Collapse
|
12
|
Webb EM, Compton A, Rai P, Chuong C, Paulson SL, Tu Z, Weger-Lucarelli J. Expression of anti-chikungunya single-domain antibodies in transgenic Aedes aegypti reduces vector competence for chikungunya virus and Mayaro virus. Front Microbiol 2023; 14:1189176. [PMID: 37378291 PMCID: PMC10291133 DOI: 10.3389/fmicb.2023.1189176] [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: 03/18/2023] [Accepted: 05/16/2023] [Indexed: 06/29/2023] Open
Abstract
Chikungunya virus (CHIKV) and Mayaro virus (MAYV) are closely related alphaviruses that cause acute febrile illness accompanied by an incapacitating polyarthralgia that can persist for years following initial infection. In conjunction with sporadic outbreaks throughout the sub-tropical regions of the Americas, increased global travel to CHIKV- and MAYV-endemic areas has resulted in imported cases of MAYV, as well as imported cases and autochthonous transmission of CHIKV, within the United States and Europe. With increasing prevalence of CHIKV worldwide and MAYV throughout the Americas within the last decade, a heavy focus has been placed on control and prevention programs. To date, the most effective means of controlling the spread of these viruses is through mosquito control programs. However, current programs have limitations in their effectiveness; therefore, novel approaches are necessary to control the spread of these crippling pathogens and lessen their disease burden. We have previously identified and characterized an anti-CHIKV single-domain antibody (sdAb) that potently neutralizes several alphaviruses including Ross River virus and Mayaro virus. Given the close antigenic relationship between MAYV and CHIKV, we formulated a single defense strategy to combat both emerging arboviruses: we generated transgenic Aedes aegypti mosquitoes that express two camelid-derived anti-CHIKV sdAbs. Following an infectious bloodmeal, we observed significant reduction in CHIKV and MAYV replication and transmission potential in sdAb-expressing transgenic compared to wild-type mosquitoes; thus, this strategy provides a novel approach to controlling and preventing outbreaks of these pathogens that reduce quality of life throughout the tropical regions of the world.
Collapse
Affiliation(s)
- Emily M. Webb
- Department of Entomology, Fralin Life Sciences Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Austin Compton
- Department of Biochemistry, Fralin Life Sciences Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Pallavi Rai
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Christina Chuong
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Sally L. Paulson
- Department of Entomology, Fralin Life Sciences Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Zhijian Tu
- Department of Biochemistry, Fralin Life Sciences Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- Center for Emerging, Zoonotic and Arthropod-Borne Pathogens, Fralin Life Sciences Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - James Weger-Lucarelli
- Department of Entomology, Fralin Life Sciences Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- Center for Emerging, Zoonotic and Arthropod-Borne Pathogens, Fralin Life Sciences Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| |
Collapse
|
13
|
Kong J, Shao G, Zhang Y, Wang J, Xie Z, Feng K, Zhang X, Xie Q. Molecular characterization, complete genome sequencing, and pathogenicity of Novel Duck Reovirus from South Coastal Area in China. Poult Sci 2023; 102:102776. [PMID: 37302330 PMCID: PMC10276289 DOI: 10.1016/j.psj.2023.102776] [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: 03/22/2023] [Accepted: 05/04/2023] [Indexed: 06/13/2023] Open
Abstract
Novel Duck Reovirus (NDRV) that has been found throughout the world in waterfowl, and it has been extensively described. Here, we report the complete genome sequence of a NDRV strain isolated in China called NDRV YF10. This strain was collected from 87 samples with infected ducks in South Coastal Area. The NDRV genome consists of 23,419 bp. With the assistance of computer analysis, the promoter and terminator of each gene segment and 10 viral genes segments were identified, which encode polypeptides ranging from 98 to 1,294 amino acids. All gene fragments of this virus strain were determined and compared to previously reported strains, revealing genetic variation with similarity rates ranging from 96 to 99% for each gene segment. Each gene segment formed 2 host-associated groups, the waterfowl-derived reovirus and the avian-derived reovirus, except for the S1 gene segment, which was closely related to ARV evolution and formed a host-independent subcluster. This difference may be due to Avian Reovirus (ARV) evolving in a host-dependent manner. In order to evaluate the pathogenicity of YF10, a novel isolated strain of NDRV was tested in 2 types of ducks. It was observed that the YF10 isolated strain exhibits varying degrees of virulence, highlighting the potential risk posed to different types of ducks. In conclusion, our findings emphasize the importance of epidemiology studies, molecular characterization, and prevention of NDRV in waterfowl.
Collapse
Affiliation(s)
- Jie Kong
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science and Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, PR China; South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, PR China; Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Guanming Shao
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science and Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, PR China; South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, PR China; Zhongshan Innovation Center of South China Agricultural University, Zhongshan 528400, PR China; Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Yukun Zhang
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science and Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, PR China; South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, PR China; Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Jinfeng Wang
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science and Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, PR China; South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, PR China; Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Zi Xie
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science and Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, PR China; South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, PR China; Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Keyu Feng
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science and Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, PR China; South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, PR China; Zhongshan Innovation Center of South China Agricultural University, Zhongshan 528400, PR China; Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, Guangzhou 510642, PR China; Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Xinheng Zhang
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science and Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, PR China; South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, PR China; Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, Guangzhou 510642, PR China; Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Qingmei Xie
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science and Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, PR China; South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, PR China; Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, Guangzhou 510642, PR China; Guangdong Provincial Key Lab of AgroAnimal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China.
| |
Collapse
|
14
|
Cereghino C, Roesch F, Carrau L, Hardy A, Ribeiro-Filho HV, Henrion-Lacritick A, Koh C, Marano JM, Bates TA, Rai P, Chuong C, Akter S, Vallet T, Blanc H, Elliott TJ, Brown AM, Michalak P, LeRoith T, Bloom JD, Marques RE, Saleh MC, Vignuzzi M, Weger-Lucarelli J. The E2 glycoprotein holds key residues for Mayaro virus adaptation to the urban Aedes aegypti mosquito. PLoS Pathog 2023; 19:e1010491. [PMID: 37018377 PMCID: PMC10109513 DOI: 10.1371/journal.ppat.1010491] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/17/2023] [Accepted: 03/13/2023] [Indexed: 04/06/2023] Open
Abstract
Adaptation to mosquito vectors suited for transmission in urban settings is a major driver in the emergence of arboviruses. To better anticipate future emergence events, it is crucial to assess their potential to adapt to new vector hosts. In this work, we used two different experimental evolution approaches to study the adaptation process of an emerging alphavirus, Mayaro virus (MAYV), to Ae. aegypti, an urban mosquito vector of many other arboviruses. We identified E2-T179N as a key mutation increasing MAYV replication in insect cells and enhancing transmission after escaping the midgut of live Ae. aegypti. In contrast, this mutation decreased viral replication and binding in human fibroblasts, a primary cellular target of MAYV in humans. We also showed that MAYV E2-T179N generates reduced viremia and displays less severe tissue pathology in vivo in a mouse model. We found evidence in mouse fibroblasts that MAYV E2-T179N is less dependent on the Mxra8 receptor for replication than WT MAYV. Similarly, exogenous expression of human apolipoprotein receptor 2 and Mxra8 enhanced WT MAYV replication compared to MAYV E2-T179N. When this mutation was introduced in the closely related chikungunya virus, which has caused major outbreaks globally in the past two decades, we observed increased replication in both human and insect cells, suggesting E2 position 179 is an important determinant of alphavirus host-adaptation, although in a virus-specific manner. Collectively, these results indicate that adaptation at the T179 residue in MAYV E2 may result in increased vector competence-but coming at the cost of optimal replication in humans-and may represent a first step towards a future emergence event.
Collapse
Affiliation(s)
- Chelsea Cereghino
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Ferdinand Roesch
- Institut Pasteur, Viral Populations and Pathogenesis Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
- UMR 1282 ISP, INRAE Centre Val de Loire, Nouzilly, France
| | - Lucía Carrau
- Institut Pasteur, Viral Populations and Pathogenesis Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
- Department of Microbiology, New York University Langone Medical Center, New York, New York, United States of America
| | - Alexandra Hardy
- Institut Pasteur, Viral Populations and Pathogenesis Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Helder V. Ribeiro-Filho
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
| | - Annabelle Henrion-Lacritick
- Institut Pasteur, Viruses and RNA Interference Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Cassandra Koh
- Institut Pasteur, Viruses and RNA Interference Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Jeffrey M. Marano
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
- Translational Biology, Medicine, and Health Graduate Program, Virginia Tech, Roanoke, Virginia, United States of America
| | - Tyler A. Bates
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Pallavi Rai
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Christina Chuong
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Shamima Akter
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
- Department of Bioinformatics and Computational Biology, School of Systems Biology, George Mason University, Fairfax, Virginia, United States of America
| | - Thomas Vallet
- Institut Pasteur, Viral Populations and Pathogenesis Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Hervé Blanc
- Institut Pasteur, Viruses and RNA Interference Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Truitt J. Elliott
- Program in Genetics, Bioinformatics, and Computational Biology (GBCB), Virginia Tech, Blacksburg, Virginia, United States of America
- Research and Informatics, University Libraries, Virginia Tech, Blacksburg, Virginia, United States of America
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Anne M. Brown
- Program in Genetics, Bioinformatics, and Computational Biology (GBCB), Virginia Tech, Blacksburg, Virginia, United States of America
| | - Pawel Michalak
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
- Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States of America
- Center for One Health Research, VA-MD Regional College of Veterinary Medicine, Blacksburg, Virginia, Untied States of Ameria
- Institute of Evolution, University of Haifa, Haifa, Israel
| | - Tanya LeRoith
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Jesse D. Bloom
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Rafael Elias Marques
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
| | - Maria-Carla Saleh
- Institut Pasteur, Viruses and RNA Interference Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Marco Vignuzzi
- Institut Pasteur, Viral Populations and Pathogenesis Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
- Institut Pasteur, Viral Populations and Pathogenesis Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| |
Collapse
|
15
|
Abbo SR, Nguyen W, Abma-Henkens MHC, van de Kamer D, Savelkoul NHA, Geertsema C, Le TTT, Tang B, Yan K, Dumenil T, van Oers MM, Suhrbier A, Pijlman GP. Comparative Efficacy of Mayaro Virus-Like Particle Vaccines Produced in Insect or Mammalian Cells. J Virol 2023; 97:e0160122. [PMID: 36883812 PMCID: PMC10062127 DOI: 10.1128/jvi.01601-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 02/13/2023] [Indexed: 03/09/2023] Open
Abstract
Mayaro virus (MAYV) is a mosquito-transmitted alphavirus that causes often debilitating rheumatic disease in tropical Central and South America. There are currently no licensed vaccines or antiviral drugs available for MAYV disease. Here, we generated Mayaro virus-like particles (VLPs) using the scalable baculovirus-insect cell expression system. High-level secretion of MAYV VLPs in the culture fluid of Sf9 insect cells was achieved, and particles with a diameter of 64 to 70 nm were obtained after purification. We characterize a C57BL/6J adult wild-type mouse model of MAYV infection and disease and used this model to compare the immunogenicity of VLPs from insect cells with that of VLPs produced in mammalian cells. Mice received two intramuscular immunizations with 1 μg of nonadjuvanted MAYV VLPs. Potent neutralizing antibody responses were generated against the vaccine strain, BeH407, with comparable activity seen against a contemporary 2018 isolate from Brazil (BR-18), whereas neutralizing activity against chikungunya virus was marginal. Sequencing of BR-18 illustrated that this virus segregates with genotype D isolates, whereas MAYV BeH407 belongs to genotype L. The mammalian cell-derived VLPs induced higher mean neutralizing antibody titers than those produced in insect cells. Both VLP vaccines completely protected adult wild-type mice against viremia, myositis, tendonitis, and joint inflammation after MAYV challenge. IMPORTANCE Mayaro virus (MAYV) is associated with acute rheumatic disease that can be debilitating and can evolve into months of chronic arthralgia. MAYV is believed to have the potential to emerge as a tropical public health threat, especially if it develops the ability to be efficiently transmitted by urban mosquito vectors, such as Aedes aegypti and/or Aedes albopictus. Here, we describe a scalable virus-like particle vaccine against MAYV that induced neutralizing antibodies against a historical and a contemporary isolate of MAYV and protected mice against infection and disease, providing a potential new intervention for MAYV epidemic preparedness.
Collapse
Affiliation(s)
- Sandra R. Abbo
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Wilson Nguyen
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Denise van de Kamer
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Niek H. A. Savelkoul
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Corinne Geertsema
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Thuy T. T. Le
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Bing Tang
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kexin Yan
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Troy Dumenil
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Monique M. van Oers
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Andreas Suhrbier
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- GVN Center of Excellence, Australian Infectious Disease Research Center, Brisbane, Queensland, Australia
| | - Gorben P. Pijlman
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| |
Collapse
|
16
|
Porier DL, Adam A, Kang L, Michalak P, Tupik J, Santos MA, Lee C, Allen IC, Wang T, Auguste AJ. Humoral and T-cell-mediated responses to a pre-clinical Zika vaccine candidate that utilizes a unique insect-specific flavivirus platform. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530296. [PMID: 36909623 PMCID: PMC10002724 DOI: 10.1101/2023.03.01.530296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Vaccination is critical for the control and prevention of viral outbreaks, yet conventional vaccine platforms may involve trade-offs between immunogenicity and safety. Insect-specific viruses have emerged as a novel vaccine platform to overcome this challenge. Detailed studies of humoral and T-cell responses induced by new insect-specific flavivirus (ISFV)-based vaccine platforms are needed to better understand correlates of protection and improve vaccine efficacy. Previously, we used a novel ISFV called Aripo virus (ARPV) to create a Zika virus (ZIKV) vaccine candidate (designated ARPV/ZIKV). ARPV/ZIKV demonstrated exceptional safety and single-dose efficacy, completely protecting mice from a lethal ZIKV challenge. Here, we explore the development of immune responses induced by ARPV/ZIKV immunization and evaluate its correlates of protection. Passive transfer of ARPV/ZIKV-induced immune sera to naïve mice prior to challenge emphasized the importance of neutralizing antibodies as a correlate of protection. Depletion of T-cells in vaccinated mice and adoptive transfer of ARPV/ZIKV-primed T-cells to naïve mice prior to challenge indicated that ARPV/ZIKV-induced CD4 + and CD8 + T-cell responses contribute to the observed protection but may not be essential for protection during ZIKV challenge. However, vaccination of Rag1 KO, Tcra KO, and muMt - mice demonstrated the critical role for ARPV/ZIKV-induced T-cells in developing protective immune responses following vaccination. Overall, both humoral and T-cell-mediated responses induced by ISFV-based vaccines are important for comprehensive immunity, and ISFV platforms continue to be a promising method for future vaccine development.
Collapse
|
17
|
Powers JM, Lyski ZL, Weber WC, Denton M, Streblow MM, Mayo AT, Haese NN, Nix CD, Rodríguez-Santiago R, Alvarado LI, Rivera-Amill V, Messer WB, Streblow DN. Infection with chikungunya virus confers heterotypic cross-neutralizing antibodies and memory B-cells against other arthritogenic alphaviruses predominantly through the B domain of the E2 glycoprotein. PLoS Negl Trop Dis 2023; 17:e0011154. [PMID: 36913428 PMCID: PMC10036167 DOI: 10.1371/journal.pntd.0011154] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 03/23/2023] [Accepted: 02/09/2023] [Indexed: 03/14/2023] Open
Abstract
Infections with Chikungunya virus, a mosquito-borne alphavirus, cause an acute febrile syndrome often followed by chronic arthritis that persists for months to years post-infection. Neutralizing antibodies are the primary immune correlate of protection elicited by infection, and the major goal of vaccinations in development. Using convalescent blood samples collected from both endemic and non-endemic human subjects at multiple timepoints following suspected or confirmed chikungunya infection, we identified antibodies with broad neutralizing properties against other alphaviruses within the Semliki Forest complex. Cross-neutralization generally did not extend to the Venezuelan Equine Encephalitis virus (VEEV) complex, although some subjects had low levels of VEEV-neutralizing antibodies. This suggests that broadly neutralizing antibodies elicited following natural infection are largely complex restricted. In addition to serology, we also performed memory B-cell analysis, finding chikungunya-specific memory B-cells in all subjects in this study as remotely as 24 years post-infection. We functionally assessed the ability of memory B-cell derived antibodies to bind to chikungunya virus, and related Mayaro virus, as well as the highly conserved B domain of the E2 glycoprotein thought to contribute to cross-reactivity between related Old-World alphaviruses. To specifically assess the role of the E2 B domain in cross-neutralization, we depleted Mayaro and Chikungunya virus E2 B domain specific antibodies from convalescent sera, finding E2B depletion significantly decreases Mayaro virus specific cross-neutralizing antibody titers with no significant effect on chikungunya virus neutralization, indicating that the E2 B domain is a key target of cross-neutralizing and potentially cross-protective neutralizing antibodies.
Collapse
Affiliation(s)
- John M. Powers
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Zoe L. Lyski
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Whitney C. Weber
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Michael Denton
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Magdalene M. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Adam T. Mayo
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Nicole N. Haese
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Chad D. Nix
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
| | | | - Luisa I. Alvarado
- Ponce Health Sciences University/ Ponce Research Institute, Ponce, Puerto Rico
| | | | - William B. Messer
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, United States of America
- Department of Medicine, Division of Infectious Disease Oregon Health and Science University, Portland, Oregon, United States of America
- OHSU-PSU School of Public Health, Program in Epidemiology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Daniel N. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| |
Collapse
|
18
|
Ramjag A, Cutrone S, Lu K, Crasto C, Jin J, Bakkour S, Carrington CVF, Simmons G. A high-throughput screening assay to identify inhibitory antibodies targeting alphavirus release. Virol J 2022; 19:170. [PMID: 36309730 PMCID: PMC9617529 DOI: 10.1186/s12985-022-01906-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Several studies have demonstrated neutralizing antibodies to be highly effective against alphavirus infection in animal models, both prophylactically and remedially. In most studies, neutralizing antibodies have been evaluated for their ability to block viral entry in vitro but recent evidence suggests that antibody inhibition through other mechanisms, including viral budding/release, significantly contributes to viral control in vivo for a number of alphaviruses. RESULTS We describe a BSL-2, cell-based, high-throughput screening system that specifically screens for inhibitors of alphavirus egress using chikungunya virus (CHIKV) and Mayaro virus (MAYV) novel replication competent nano-luciferase (nLuc) reporter viruses. Screening of both polyclonal sera and memory B-cell clones from CHIKV immune individuals using the optimized assay detected several antibodies that display potent anti-budding activity. CONCLUSIONS We describe an "anti-budding assay" to specifically screen for inhibitors of viral egress using novel CHIKV and MAYV nLuc reporter viruses. This BSL-2 safe, high-throughput system can be utilized to explore neutralizing "anti-budding" antibodies to yield potent candidates for CHIKV and MAYV therapeutics and prophylaxis.
Collapse
Affiliation(s)
- Anushka Ramjag
- Department of Preclinical Sciences, The University of the West Indies, St. Augustine Campus, St. Augustine, Trinidad and Tobago
| | - Sergej Cutrone
- Vitalant Research Institute, 270 Masonic Avenue, San Francisco, CA, 94118, USA
- University of California San Francisco, San Francisco, CA, 94143, USA
| | - Kai Lu
- Vitalant Research Institute, 270 Masonic Avenue, San Francisco, CA, 94118, USA
| | - Christine Crasto
- Vitalant Research Institute, 270 Masonic Avenue, San Francisco, CA, 94118, USA
| | - Jing Jin
- Vitalant Research Institute, 270 Masonic Avenue, San Francisco, CA, 94118, USA
- University of California San Francisco, San Francisco, CA, 94143, USA
| | - Sonia Bakkour
- Vitalant Research Institute, 270 Masonic Avenue, San Francisco, CA, 94118, USA
- University of California San Francisco, San Francisco, CA, 94143, USA
| | - Christine V F Carrington
- Department of Preclinical Sciences, The University of the West Indies, St. Augustine Campus, St. Augustine, Trinidad and Tobago
| | - Graham Simmons
- Vitalant Research Institute, 270 Masonic Avenue, San Francisco, CA, 94118, USA.
- University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
19
|
A Bivalent Trans-Amplifying RNA Vaccine Candidate Induces Potent Chikungunya and Ross River Virus Specific Immune Responses. Vaccines (Basel) 2022; 10:vaccines10091374. [PMID: 36146452 PMCID: PMC9503900 DOI: 10.3390/vaccines10091374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/17/2022] Open
Abstract
Alphaviruses such as the human pathogenic chikungunya virus (CHIKV) and Ross River virus (RRV) can cause explosive outbreaks raising public health concerns. However, no vaccine or specific antiviral treatment is yet available. We recently established a CHIKV vaccine candidate based on trans-amplifying RNA (taRNA). This novel system consists of a replicase-encoding mRNA and a trans-replicon (TR) RNA encoding the antigen. The TR-RNA is amplified by the replicase in situ. We were interested in determining whether multiple TR-RNAs can be amplified in parallel and if, thus, a multivalent vaccine candidate can be generated. In vitro, we observed an efficient amplification of two TR-RNAs, encoding for the CHIKV and the RRV envelope proteins, by the replicase, which resulted in a high antigen expression. Vaccination of BALB/c mice with the two TR-RNAs induced CHIKV- and RRV-specific humoral and cellular immune responses. However, antibody titers and neutralization capacity were higher after immunization with a single TR-RNA. In contrast, alphavirus-specific T cell responses were equally potent after the bivalent vaccination. These data show the proof-of-principle that the taRNA system can be used to generate multivalent vaccines; however, further optimizations will be needed for clinical application.
Collapse
|
20
|
Andreolla AP, Borges AA, Bordignon J, Duarte dos Santos CN. Mayaro Virus: The State-of-the-Art for Antiviral Drug Development. Viruses 2022; 14:1787. [PMID: 36016409 PMCID: PMC9415492 DOI: 10.3390/v14081787] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 12/18/2022] Open
Abstract
Mayaro virus is an emerging arbovirus that causes nonspecific febrile illness or arthralgia syndromes similar to the Chikungunya virus, a virus closely related from the Togaviridae family. MAYV outbreaks occur more frequently in the northern and central-western states of Brazil; however, in recent years, virus circulation has been spreading to other regions. Due to the undifferentiated initial clinical symptoms between MAYV and other endemic pathogenic arboviruses with geographic overlapping, identification of patients infected by MAYV might be underreported. Additionally, the lack of specific prophylactic approaches or antiviral drugs limits the pharmacological management of patients to treat symptoms like pain and inflammation, as is the case with most pathogenic alphaviruses. In this context, this review aims to present the state-of-the-art regarding the screening and development of compounds/molecules which may present anti-MAYV activity and infection inhibition.
Collapse
Affiliation(s)
- Ana Paula Andreolla
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, ICC/Fiocruz, Curitiba 81350-010, PR, Brazil
- Departamento de Biologia Celular e Molecular, Universidade Federal do Paraná, Curitiba 81530-900, PR, Brazil
| | - Alessandra Abel Borges
- Laboratório de Pesquisas em Virologia e Imunologia, Universidade Federal de Alagoas, Maceió 57072-900, AL, Brazil
| | - Juliano Bordignon
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, ICC/Fiocruz, Curitiba 81350-010, PR, Brazil
| | | |
Collapse
|
21
|
Slifka DK, Raué HP, Weber WC, Andoh TF, Kreklywich CN, DeFilippis VR, Streblow DN, Slifka MK, Amanna IJ. Development of a next-generation chikungunya virus vaccine based on the HydroVax platform. PLoS Pathog 2022; 18:e1010695. [PMID: 35788221 PMCID: PMC9286250 DOI: 10.1371/journal.ppat.1010695] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 07/15/2022] [Accepted: 06/22/2022] [Indexed: 12/02/2022] Open
Abstract
Chikungunya virus (CHIKV) is an emerging/re-emerging mosquito-borne pathogen responsible for explosive epidemics of febrile illness characterized by debilitating polyarthralgia and the risk of lethal infection among the most severe cases. Despite the public health risk posed by CHIKV, no vaccine is currently available. Using a site-directed hydrogen peroxide-based inactivation approach, we developed a new CHIKV vaccine, HydroVax-CHIKV. This vaccine technology was compared to other common virus inactivation approaches including β-propiolactone (BPL), formaldehyde, heat, and ultraviolet (UV) irradiation. Heat, UV, and BPL were efficient at inactivating CHIKV-181/25 but caused substantial damage to neutralizing epitopes and failed to induce high-titer neutralizing antibodies in vaccinated mice. HydroVax-CHIKV and formaldehyde-inactivated CHIKV retained intact neutralizing epitopes similar to live virus controls but the HydroVax-CHIKV approach demonstrated a more rapid rate of virus inactivation. HydroVax-CHIKV vaccination induced high neutralizing responses to homologous and heterologous CHIKV clades as well as to other alphaviruses including Mayaro virus, O'nyong'nyong virus, and Una virus. Following heterologous infection with CHIKV-SL15649, HydroVax-CHIKV-immunized mice were protected against viremia, CHIKV-associated arthritic disease, and lethal CHIKV infection by an antibody-dependent mechanism. In contrast, animals vaccinated with Heat- or UV-inactivated virus showed no protection against viremia in addition to demonstrating significantly exacerbated CD4+ T cell-mediated footpad swelling after CHIKV infection. Together, these results demonstrate the risks associated with using suboptimal inactivation methods that fail to elicit protective neutralizing antibody responses and show that HydroVax-CHIKV represents a promising new vaccine candidate for prevention of CHIKV-associated disease.
Collapse
Affiliation(s)
- Dawn K. Slifka
- Najít Technologies, Incorporated, Beaverton, Oregon, United States of America
| | - Hans-Peter Raué
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Whitney C. Weber
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Takeshi F. Andoh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Craig N. Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Victor R. DeFilippis
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Daniel N. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Mark K. Slifka
- Najít Technologies, Incorporated, Beaverton, Oregon, United States of America
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Ian J. Amanna
- Najít Technologies, Incorporated, Beaverton, Oregon, United States of America
| |
Collapse
|
22
|
Kafai NM, Diamond MS, Fox JM. Distinct Cellular Tropism and Immune Responses to Alphavirus Infection. Annu Rev Immunol 2022; 40:615-649. [PMID: 35134315 DOI: 10.1146/annurev-immunol-101220-014952] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Alphaviruses are emerging and reemerging viruses that cause disease syndromes ranging from incapacitating arthritis to potentially fatal encephalitis. While infection by arthritogenic and encephalitic alphaviruses results in distinct clinical manifestations, both virus groups induce robust innate and adaptive immune responses. However, differences in cellular tropism, type I interferon induction, immune cell recruitment, and B and T cell responses result in differential disease progression and outcome. In this review, we discuss aspects of immune responses that contribute to protective or pathogenic outcomes after alphavirus infection. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Natasha M Kafai
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA; , .,Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Michael S Diamond
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA; , .,Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA.,Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Julie M Fox
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA;
| |
Collapse
|
23
|
Bopp NE, Jencks KJ, Siles C, Guevara C, Vilcarromero S, Fernández D, Halsey ES, Ampuero JS, Aguilar PV. Serological Responses in Patients Infected with Mayaro Virus and Evaluation of Cross-Protective Responses against Chikungunya Virus. Am J Trop Med Hyg 2022; 106:607-609. [PMID: 34844213 PMCID: PMC8832936 DOI: 10.4269/ajtmh.21-0579] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/05/2021] [Indexed: 02/03/2023] Open
Abstract
Mayaro virus (MAYV) is an alphavirus endemic to both Latin America and the Caribbean. Recent reports have questioned the ability of MAYV and its close relative, Chikungunya virus (CHIKV), to generate cross-reactive, neutralizing antibodies to one another. Since CHIKV was introduced to South America in 2013, discerning whether individuals have cross-reactive antibodies or whether they have had exposures to both viruses previously has been difficult. Using samples obtained from people infected with MAYV prior to the introduction of CHIKV in the Americas, we performed neutralizing assays and observed no discernable neutralization of CHIKV by sera from patients previously infected with MAYV. These data suggest that a positive CHIKV neutralization test cannot be attributed to prior exposure to MAYV and that previous exposure to MAYV may not be protective against a subsequent CHIKV infection.
Collapse
Affiliation(s)
- Nathen E. Bopp
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| | - Kara J. Jencks
- School of Medicine, University of Texas Medical Branch, Galveston, Texas
| | | | | | | | - Diana Fernández
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas;,Center for Tropical Diseases, University of Texas Medical Branch, Galveston, Texas
| | | | | | - Patricia V. Aguilar
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas;,Center for Tropical Diseases, University of Texas Medical Branch, Galveston, Texas;,Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas,Address correspondence to Patricia V. Aguilar, Department of Pathology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0609. E-mail:
| |
Collapse
|
24
|
Chikungunya Virus Exposure Partially Cross-Protects against Mayaro Virus Infection in Mice. J Virol 2021; 95:e0112221. [PMID: 34549980 DOI: 10.1128/jvi.01122-21] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Chikungunya virus (CHIKV) and Mayaro virus (MAYV) are closely related members of the Semliki Forest virus antigenic complex classified as belonging to the genus Alphavirus of the family Togaviridae. These viruses cause human disease, with sudden fever and joint inflammation that can persist for long periods. CHIKV is the causative agent of large outbreaks worldwide, and MAYV infection represents a growing public health concern in Latin America, causing sporadic cases and geographically limited outbreaks. Considering the relationship between CHIKV and MAYV, the present study aimed to evaluate if preexisting CHIKV immunity protects against MAYV infection. Immunocompetent C57BL/6 mice were intraperitoneally infected with CHIKV and, 4 weeks later, they were infected with MAYV in their hind paw. We observed that the preexistence of CHIKV immunity conferred partial cross-protection against secondary MAYV infection, reducing disease severity, tissue viral load, and histopathological scores. Interestingly, CHIKV antibodies from humans and mice showed low cross-neutralization to MAYV, but neutralizing activity significantly increased after secondary infection. Furthermore, depletion of adaptive immune cells (CD4+ T, CD8+ T, and CD19+ B cells) did not alter the cross-protection phenotype, suggesting that distinct cell subsets or a combination of adaptive immune cells stimulated by CHIKV are responsible for the partial cross-protection against MAYV. The reduction of proinflammatory cytokines, such as interferon gamma (IFN-γ), in animals secondarily infected by MAYV, suggests a role for innate immunity in cross-protection. Our findings shed light on how preexisting immunity to arthritogenic alphaviruses may affect secondary infection, which may further develop relevant influence in disease outcome and viral transmission. IMPORTANCE Mosquito-borne viruses have a worldwide impact, especially in tropical climates. Chikungunya virus has been present mostly in developing countries, causing millions of infections, while Mayaro virus, a close relative, has been limited to the Caribbean and tropical regions of Latin America. The potential emergence and spread of Mayaro virus to other high-risk areas have increased the scientific community's attention to an imminent worldwide epidemic. Here, we designed an experimental protocol of chikungunya and Mayaro virus mouse infection, which develops a measurable and quantifiable disease that allows us to make inferences about potential immunological effects during secondary virus infection. Our results demonstrate that previous chikungunya virus infection is able to reduce the severity of clinical outcomes during secondary Mayaro infection. We provide scientific understanding of immunological features during secondary infection with the closely related virus, thus assisting in better comprehending viral transmission and the pathological outcome of these diseases.
Collapse
|
25
|
Near-germline human monoclonal antibodies neutralize and protect against multiple arthritogenic alphaviruses. Proc Natl Acad Sci U S A 2021; 118:2100104118. [PMID: 34507983 DOI: 10.1073/pnas.2100104118] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2021] [Indexed: 12/11/2022] Open
Abstract
Arthritogenic alphaviruses are globally distributed, mosquito-transmitted viruses that cause rheumatological disease in humans and include Chikungunya virus (CHIKV), Mayaro virus (MAYV), and others. Although serological evidence suggests that some antibody-mediated heterologous immunity may be afforded by alphavirus infection, the extent to which broadly neutralizing antibodies that protect against multiple arthritogenic alphaviruses are elicited during natural infection remains unknown. Here, we describe the isolation and characterization of MAYV-reactive alphavirus monoclonal antibodies (mAbs) from a CHIKV-convalescent donor. We characterized 33 human mAbs that cross-reacted with CHIKV and MAYV and engaged multiple epitopes on the E1 and E2 glycoproteins. We identified five mAbs that target distinct regions of the B domain of E2 and potently neutralize multiple alphaviruses with differential breadth of inhibition. These broadly neutralizing mAbs (bNAbs) contain few somatic mutations and inferred germline-revertants retained neutralizing capacity. Two bNAbs, DC2.M16 and DC2.M357, protected against both CHIKV- and MAYV-induced musculoskeletal disease in mice. These findings enhance our understanding of the cross-reactive and cross-protective antibody response to human alphavirus infections.
Collapse
|
26
|
Kim AS, Kafai NM, Winkler ES, Gilliland TC, Cottle EL, Earnest JT, Jethva PN, Kaplonek P, Shah AP, Fong RH, Davidson E, Malonis RJ, Quiroz JA, Williamson LE, Vang L, Mack M, Crowe JE, Doranz BJ, Lai JR, Alter G, Gross ML, Klimstra WB, Fremont DH, Diamond MS. Pan-protective anti-alphavirus human antibodies target a conserved E1 protein epitope. Cell 2021; 184:4414-4429.e19. [PMID: 34416146 PMCID: PMC8382027 DOI: 10.1016/j.cell.2021.07.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 04/01/2021] [Accepted: 07/02/2021] [Indexed: 12/13/2022]
Abstract
Alphaviruses are emerging, mosquito-transmitted pathogens that cause musculoskeletal and neurological disease in humans. Although neutralizing antibodies that inhibit individual alphaviruses have been described, broadly reactive antibodies that protect against both arthritogenic and encephalitic alphaviruses have not been reported. Here, we identify DC2.112 and DC2.315, two pan-protective yet poorly neutralizing human monoclonal antibodies (mAbs) that avidly bind to viral antigen on the surface of cells infected with arthritogenic and encephalitic alphaviruses. These mAbs engage a conserved epitope in domain II of the E1 protein proximal to and within the fusion peptide. Treatment with DC2.112 or DC2.315 protects mice against infection by both arthritogenic (chikungunya and Mayaro) and encephalitic (Venezuelan, Eastern, and Western equine encephalitis) alphaviruses through multiple mechanisms, including inhibition of viral egress and monocyte-dependent Fc effector functions. These findings define a conserved epitope recognized by weakly neutralizing yet protective antibodies that could be targeted for pan-alphavirus immunotherapy and vaccine design.
Collapse
Affiliation(s)
- Arthur S Kim
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Natasha M Kafai
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Emma S Winkler
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Theron C Gilliland
- Center for Vaccine Research and Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Emily L Cottle
- Center for Vaccine Research and Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - James T Earnest
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Prashant N Jethva
- Department of Chemistry, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Paulina Kaplonek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Aadit P Shah
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Rachel H Fong
- Integral Molecular, Inc., Philadelphia, PA 19104, USA
| | | | - Ryan J Malonis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jose A Quiroz
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Lauren E Williamson
- Vanderbilt Vaccine Center and Departments of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lo Vang
- Emergent BioSolutions, Gaithersburg, MD 20879, USA
| | - Matthias Mack
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - James E Crowe
- Vanderbilt Vaccine Center and Departments of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | - Jonathan R Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Michael L Gross
- Department of Chemistry, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - William B Klimstra
- Center for Vaccine Research and Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO 63110, USA; Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
27
|
Napoleão-Pêgo P, Carneiro FRG, Durans AM, Gomes LR, Morel CM, Provance DW, De-Simone SG. Performance assessment of a multi-epitope chimeric antigen for the serological diagnosis of acute Mayaro fever. Sci Rep 2021; 11:15374. [PMID: 34321560 PMCID: PMC8319364 DOI: 10.1038/s41598-021-94817-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 07/12/2021] [Indexed: 02/04/2023] Open
Abstract
Mayaro virus (MAYV), which causes mayaro fever, is endemic to limited regions of South America that may expand due to the possible involvement of Aedes spp. mosquitoes in its transmission. Its effective control will require the accurate identification of infected individuals, which has been restricted to nucleic acid-based tests due to similarities with other emerging members of the Alphavirus genus of the Togaviridae family; both in structure and clinical symptoms. Serological tests have a more significant potential to expand testing at a reasonable cost, and their performance primarily reflects that of the antigen utilized to capture pathogen-specific antibodies. Here, we describe the assembly of a synthetic gene encoding multiple copies of antigenic determinants mapped from the nsP1, nsP2, E1, and E2 proteins of MAYV that readily expressed as a stable chimeric protein in bacteria. Its serological performance as the target in ELISAs revealed a high accuracy for detecting anti-MAYV IgM antibodies. No cross-reactivity was observed with serum from seropositive individuals for dengue, chikungunya, yellow fever, Zika, and other infectious diseases as well as healthy individuals. Our data suggest that this bioengineered antigen could be used to develop high-performance serological tests for MAYV infections.
Collapse
Affiliation(s)
- Paloma Napoleão-Pêgo
- Oswaldo Cruz Foundation (FIOCRUZ), Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Diseases of Neglected Populations (INCT-IDPN), Brazil Av 4365, Leonidas Deane Building, Room 309, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Flávia R G Carneiro
- Oswaldo Cruz Foundation (FIOCRUZ), Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Diseases of Neglected Populations (INCT-IDPN), Brazil Av 4365, Leonidas Deane Building, Room 309, Rio de Janeiro, RJ, 21040-900, Brazil.,Laboratory of Interdisplinary Medical Research (LIPMED), Oswaldo Cruz Institute (IOC), FIOCRUZ, Brazil Av 4365, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Andressa M Durans
- Oswaldo Cruz Foundation (FIOCRUZ), Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Diseases of Neglected Populations (INCT-IDPN), Brazil Av 4365, Leonidas Deane Building, Room 309, Rio de Janeiro, RJ, 21040-900, Brazil.,Laboratory of Interdisplinary Medical Research (LIPMED), Oswaldo Cruz Institute (IOC), FIOCRUZ, Brazil Av 4365, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Larissa R Gomes
- Oswaldo Cruz Foundation (FIOCRUZ), Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Diseases of Neglected Populations (INCT-IDPN), Brazil Av 4365, Leonidas Deane Building, Room 309, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Carlos M Morel
- Oswaldo Cruz Foundation (FIOCRUZ), Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Diseases of Neglected Populations (INCT-IDPN), Brazil Av 4365, Leonidas Deane Building, Room 309, Rio de Janeiro, RJ, 21040-900, Brazil
| | - David W Provance
- Oswaldo Cruz Foundation (FIOCRUZ), Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Diseases of Neglected Populations (INCT-IDPN), Brazil Av 4365, Leonidas Deane Building, Room 309, Rio de Janeiro, RJ, 21040-900, Brazil.,Laboratory of Interdisplinary Medical Research (LIPMED), Oswaldo Cruz Institute (IOC), FIOCRUZ, Brazil Av 4365, Rio de Janeiro, RJ, 21040-900, Brazil
| | - Salvatore G De-Simone
- Oswaldo Cruz Foundation (FIOCRUZ), Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Diseases of Neglected Populations (INCT-IDPN), Brazil Av 4365, Leonidas Deane Building, Room 309, Rio de Janeiro, RJ, 21040-900, Brazil. .,Biology Institute, Federal Fluminense University, Outeiro de São Joao Batista S/N, Niterói, RJ, 24020-141, Brazil.
| |
Collapse
|
28
|
Auguste AJ, Langsjoen RM, Porier DL, Erasmus JH, Bergren NA, Bolling BG, Luo H, Singh A, Guzman H, Popov VL, Travassos da Rosa APA, Wang T, Kang L, Allen IC, Carrington CVF, Tesh RB, Weaver SC. Isolation of a novel insect-specific flavivirus with immunomodulatory effects in vertebrate systems. Virology 2021; 562:50-62. [PMID: 34256244 DOI: 10.1016/j.virol.2021.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/03/2021] [Accepted: 07/05/2021] [Indexed: 12/13/2022]
Abstract
We describe the isolation and characterization of a novel insect-specific flavivirus (ISFV), tentatively named Aripo virus (ARPV), that was isolated from Psorophora albipes mosquitoes collected in Trinidad. The ARPV genome was determined and phylogenetic analyses showed that it is a dual host associated ISFV, and clusters with the main mosquito-borne flaviviruses. ARPV antigen was significantly cross-reactive with Japanese encephalitis virus serogroup antisera, with significant cross-reactivity to Ilheus and West Nile virus (WNV). Results suggest that ARPV replication is limited to mosquitoes, as it did not replicate in the sandfly, culicoides or vertebrate cell lines tested. We also demonstrated that ARPV is endocytosed into vertebrate cells and is highly immunomodulatory, producing a robust innate immune response despite its inability to replicate in vertebrate systems. We show that prior infection or coinfection with ARPV limits WNV-induced disease in mouse models, likely the result of a robust ARPV-induced type I interferon response.
Collapse
Affiliation(s)
- Albert J Auguste
- Department of Entomology, College of Agriculture and Life Sciences, Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA; Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA.
| | - Rose M Langsjoen
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Danielle L Porier
- Department of Entomology, College of Agriculture and Life Sciences, Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Jesse H Erasmus
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Nicholas A Bergren
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Bethany G Bolling
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Huanle Luo
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ankita Singh
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Hilda Guzman
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Vsevolod L Popov
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | - Tian Wang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Lin Kang
- Edward Via College of Osteopathic Medicine, Monroe, LA, 71203, USA; Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, 24060, USA
| | - Irving C Allen
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA; Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, 24060, USA
| | - Christine V F Carrington
- Department of Preclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Robert B Tesh
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Scott C Weaver
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
29
|
Caicedo EY, Charniga K, Rueda A, Dorigatti I, Mendez Y, Hamlet A, Carrera JP, Cucunubá ZM. The epidemiology of Mayaro virus in the Americas: A systematic review and key parameter estimates for outbreak modelling. PLoS Negl Trop Dis 2021; 15:e0009418. [PMID: 34081717 PMCID: PMC8205173 DOI: 10.1371/journal.pntd.0009418] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 06/15/2021] [Accepted: 04/27/2021] [Indexed: 01/05/2023] Open
Abstract
Mayaro virus (MAYV) is an arbovirus that is endemic to tropical forests in Central and South America, particularly within the Amazon basin. In recent years, concern has increased regarding MAYV's ability to invade urban areas and cause epidemics across the region. We conducted a systematic literature review to characterise the evolutionary history of MAYV, its transmission potential, and exposure patterns to the virus. We analysed data from the literature on MAYV infection to produce estimates of key epidemiological parameters, including the generation time and the basic reproduction number, R0. We also estimated the force-of-infection (FOI) in epidemic and endemic settings. Seventy-six publications met our inclusion criteria. Evidence of MAYV infection in humans, animals, or vectors was reported in 14 Latin American countries. Nine countries reported evidence of acute infection in humans confirmed by viral isolation or reverse transcription-PCR (RT-PCR). We identified at least five MAYV outbreaks. Seroprevalence from population based cross-sectional studies ranged from 21% to 72%. The estimated mean generation time of MAYV was 15.2 days (95% CrI: 11.7-19.8) with a standard deviation of 6.3 days (95% CrI: 4.2-9.5). The per-capita risk of MAYV infection (FOI) ranged between 0.01 and 0.05 per year. The mean R0 estimates ranged between 2.1 and 2.9 in the Amazon basin areas and between 1.1 and 1.3 in the regions outside of the Amazon basin. Although MAYV has been identified in urban vectors, there is not yet evidence of sustained urban transmission. MAYV's enzootic cycle could become established in forested areas within cities similar to yellow fever virus.
Collapse
Affiliation(s)
| | - Kelly Charniga
- MRC Centre for Global Infectious Disease Analysis (MRC-GIDA), Imperial College London, London, United Kingdom
| | - Amanecer Rueda
- Universidad Pedagógica y Tecnológica de Colombia, Tunja, Colombia
| | - Ilaria Dorigatti
- MRC Centre for Global Infectious Disease Analysis (MRC-GIDA), Imperial College London, London, United Kingdom
| | - Yardany Mendez
- Universidad Pedagógica y Tecnológica de Colombia, Tunja, Colombia
| | - Arran Hamlet
- MRC Centre for Global Infectious Disease Analysis (MRC-GIDA), Imperial College London, London, United Kingdom
| | - Jean-Paul Carrera
- Department of Zoology, University of Oxford, Oxford, United Kingdom
- Department of Research in Virology and Biotechnology, Gorgas Memorial Institute of Health Studies, Panama City, Panama
| | - Zulma M. Cucunubá
- MRC Centre for Global Infectious Disease Analysis (MRC-GIDA), Imperial College London, London, United Kingdom
| |
Collapse
|
30
|
Powers JM, Haese NN, Denton M, Ando T, Kreklywich C, Bonin K, Streblow CE, Kreklywich N, Smith P, Broeckel R, DeFilippis V, Morrison TE, Heise MT, Streblow DN. Non-replicating adenovirus based Mayaro virus vaccine elicits protective immune responses and cross protects against other alphaviruses. PLoS Negl Trop Dis 2021; 15:e0009308. [PMID: 33793555 PMCID: PMC8051823 DOI: 10.1371/journal.pntd.0009308] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 04/16/2021] [Accepted: 03/15/2021] [Indexed: 11/20/2022] Open
Abstract
Mayaro virus (MAYV) is an alphavirus endemic to South and Central America associated with sporadic outbreaks in humans. MAYV infection causes severe joint and muscle pain that can persist for weeks to months. Currently, there are no approved vaccines or therapeutics to prevent MAYV infection or treat the debilitating musculoskeletal inflammatory disease. In the current study, a prophylactic MAYV vaccine expressing the complete viral structural polyprotein was developed based on a non-replicating human adenovirus V (AdV) platform. Vaccination with AdV-MAYV elicited potent neutralizing antibodies that protected WT mice against MAYV challenge by preventing viremia, reducing viral dissemination to tissues and mitigating viral disease. The vaccine also prevented viral-mediated demise in IFN⍺R1-/- mice. Passive transfer of immune serum from vaccinated animals similarly prevented infection and disease in WT mice as well as virus-induced demise of IFN⍺R1-/- mice, indicating that antiviral antibodies are protective. Immunization with AdV-MAYV also generated cross-neutralizing antibodies against two related arthritogenic alphaviruses-chikungunya and Una viruses. These cross-neutralizing antibodies were protective against lethal infection in IFN⍺R1-/- mice following challenge with these heterotypic alphaviruses. These results indicate AdV-MAYV elicits protective immune responses with substantial cross-reactivity and protective efficacy against other arthritogenic alphaviruses. Our findings also highlight the potential for development of a multi-virus targeting vaccine against alphaviruses with endemic and epidemic potential in the Americas.
Collapse
Affiliation(s)
- John M. Powers
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Nicole N. Haese
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Michael Denton
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Takeshi Ando
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Craig Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Kiley Bonin
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Cassilyn E. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Nicholas Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Patricia Smith
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Rebecca Broeckel
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Victor DeFilippis
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Thomas E. Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Mark T. Heise
- Department of Genetics, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Daniel N. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| |
Collapse
|
31
|
Hoque H, Islam R, Ghosh S, Rahaman MM, Jewel NA, Miah MA. Implementation of in silico methods to predict common epitopes for vaccine development against Chikungunya and Mayaro viruses. Heliyon 2021; 7:e06396. [PMID: 33732931 PMCID: PMC7944042 DOI: 10.1016/j.heliyon.2021.e06396] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/02/2021] [Accepted: 02/25/2021] [Indexed: 01/03/2023] Open
Abstract
Being a Positive sense RNA virus the recent reemergence of Chikungunya and Mayaro virus has taken the concern of the leading scientific communities of the world. Though the outbreak of Mayaro virus is limited to Neotropical region only, Chikungunya is already identified in over 60 countries around the world. Besides, the lack of a strong protective treatment, misdiagnosis issue and co-circulation of both the viruses calls for a new strategy which could potentially prevent these infections from spreading. In this study, we therefore, identified the peptide based vaccine candidates e.g. epitopes for B cell and T cell from Chikungunya virus which also showed to be homologous to the Mayaro virus through immuno-informatics and computational approaches. Final epitopes identified from the most antigenic structural polyprotein of both the viruses were 5 for CD8+ T cell Epitopes (209KPGDSGRPI217, 219TGTMGHFIL227, 239ALSVVTWNK247, 98KPGRRERMC106 and 100GRRERMCMK108), 2 epitopes for CD4+ T cell (105MCMKIENDCIFEVKH119 and 502DRTLLSQQSGNVKIT516) and a single epitope for B cell (504GGRFTIPTGAGKPGDSGRPI518). Analysis of our predicted epitopes for population coverage showed prominent population coverage (92.43%) around the world. Finally, molecular docking simulation of the foreseen T cell epitopes with respondent HLA alleles secured good HLA-epitope interaction. This study was directed towards the discovery of potential antigenic epitopes which can open up a new skyline to design novel vaccines for combating both of the diseases at the same time.
Collapse
Affiliation(s)
- Hammadul Hoque
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Rahatul Islam
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Srijon Ghosh
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Md. Mashiur Rahaman
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Nurnabi Azad Jewel
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Md. Abunasar Miah
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| |
Collapse
|
32
|
Campos RK, Preciado-Llanes L, Azar SR, Kim YC, Brandon O, López-Camacho C, Reyes-Sandoval A, Rossi SL. Adenoviral-Vectored Mayaro and Chikungunya Virus Vaccine Candidates Afford Partial Cross-Protection From Lethal Challenge in A129 Mouse Model. Front Immunol 2020; 11:591885. [PMID: 33224148 PMCID: PMC7672187 DOI: 10.3389/fimmu.2020.591885] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/07/2020] [Indexed: 01/08/2023] Open
Abstract
Mayaro (MAYV) and chikungunya viruses (CHIKV) are vector-borne arthritogenic alphaviruses that cause acute febrile illnesses. CHIKV is widespread and has recently caused large urban outbreaks, whereas the distribution of MAYV is restricted to tropical areas in South America with small and sporadic outbreaks. Because MAYV and CHIKV are closely related and have high amino acid similarity, we investigated whether vaccination against one could provide cross-protection against the other. We vaccinated A129 mice (IFNAR -/-) with vaccines based on chimpanzee adenoviral vectors encoding the structural proteins of either MAYV or CHIKV. ChAdOx1 May is a novel vaccine against MAYV, whereas ChAdOx1 Chik is a vaccine against CHIKV already undergoing early phase I clinical trials. We demonstrate that ChAdOx1 May was able to afford full protection against MAYV challenge in mice, with most samples yielding neutralizing PRNT80 antibody titers of 1:258. ChAdOx1 May also provided partial cross-protection against CHIKV, with protection being assessed using the following parameters: survival, weight loss, foot swelling and viremia. Reciprocally, ChAdOx1 Chik vaccination reduced MAYV viral load, as well as morbidity and lethality caused by this virus, but did not protect against foot swelling. The cross-protection observed is likely to be, at least in part, secondary to cross-neutralizing antibodies induced by both vaccines. In summary, our findings suggest that ChAdOx1 Chik and ChAdOx1 May vaccines are not only efficacious against CHIKV and MAYV, respectively, but also afford partial heterologous cross-protection.
Collapse
Affiliation(s)
- Rafael Kroon Campos
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Lorena Preciado-Llanes
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Sasha R. Azar
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Young Chan Kim
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Olivia Brandon
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - César López-Camacho
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Arturo Reyes-Sandoval
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Shannan L. Rossi
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
33
|
Carrera JP, Pittí Y, Molares-Martínez JC, Casal E, Pereyra-Elias R, Saenz L, Guerrero I, Galué J, Rodriguez-Alvarez F, Jackman C, Pascale JM, Armien B, Weaver SC, Donnelly CA, Vittor AY. Clinical and Serological Findings of Madariaga and Venezuelan Equine Encephalitis Viral Infections: A Follow-up Study 5 Years After an Outbreak in Panama. Open Forum Infect Dis 2020; 7:ofaa359. [PMID: 33005697 PMCID: PMC7518370 DOI: 10.1093/ofid/ofaa359] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/13/2020] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Human cases of Madariaga virus (MADV) infection were first detected during an outbreak in 2010 in eastern Panama, where Venezuelan equine encephalitis virus (VEEV) also circulates. Little is known about the long-term consequences of either alphavirus infection. METHODS A follow-up study of the 2010 outbreak was undertaken in 2015. An additional survey was carried out 2 weeks after a separate 2017 alphavirus outbreak in a neighboring population in eastern Panama. Serological studies and statistical analyses were undertaken in both populations. RESULTS Among the originally alphavirus-seronegative participants (n = 35 of 65), seroconversion was observed at a rate of 14.3% (95% CI, 4.8%-30.3%) for MADV and 8.6% (95% CI, 1.8%-23.1%) for VEEV over 5 years. Among the originally MADV-seropositive participants (n = 14 of 65), VEEV seroconversion occurred in 35.7% (95% CI, 12.8%-64.9%). In the VEEV-seropositive participants (n = 16 of 65), MADV seroconversion occurred in 6.3% (95% CI, 0.2%-30.2%). MADV seroreversion was observed in 14.3% (95% CI, 1.8%-42.8%) of those who were originally seropositive in 2010. VEEV seroconversion in the baseline MADV-seropositive participants was significantly higher than in alphavirus-negative participants. In the population sampled in 2017, MADV and VEEV seroprevalence was 13.2% and 16.8%, respectively. Memory loss, insomnia, irritability, and seizures were reported significantly more frequently in alphavirus-seropositive participants than in seronegative participants. CONCLUSIONS High rates of seroconversion to MADV and VEEV over 5 years suggest frequent circulation of both viruses in Panama. Enhanced susceptibility to VEEV infection may be conferred by MADV infection. We provide evidence of persistent neurologic symptoms up to 5 years following MADV and VEEV exposure.
Collapse
Affiliation(s)
- Jean-Paul Carrera
- Department of Zoology, University of Oxford, Oxford, UK
- Department of Research in Virology and Biotechnology, Gorgas Memorial Institute of Health Studies, Panama City, Panama
| | - Yaneth Pittí
- Department of Research in Virology and Biotechnology, Gorgas Memorial Institute of Health Studies, Panama City, Panama
| | - Juan C Molares-Martínez
- Department of Research in Virology and Biotechnology, Gorgas Memorial Institute of Health Studies, Panama City, Panama
| | - Eric Casal
- Department of Research in Virology and Biotechnology, Gorgas Memorial Institute of Health Studies, Panama City, Panama
| | - Reneé Pereyra-Elias
- National Perinatal Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Lisseth Saenz
- Department of Research in Virology and Biotechnology, Gorgas Memorial Institute of Health Studies, Panama City, Panama
| | - Isela Guerrero
- Department of Research in Virology and Biotechnology, Gorgas Memorial Institute of Health Studies, Panama City, Panama
| | - Josefrancisco Galué
- Department of Research in Virology and Biotechnology, Gorgas Memorial Institute of Health Studies, Panama City, Panama
| | - Fatima Rodriguez-Alvarez
- Department of Research in Virology and Biotechnology, Gorgas Memorial Institute of Health Studies, Panama City, Panama
| | - Carmela Jackman
- Department of Epidemiology, Ministry of Health, Panama, Panama
| | - Juan Miguel Pascale
- Clinical Research Unit, Gorgas Memorial Institute of Health Studies, Panama City, Panama
| | - Blas Armien
- Department of Research in Emerging and Zoonotic Diseases, Gorgas Memorial Institute of Health Studies, Panama City, Panama
| | - Scott C Weaver
- Institute for Human Infection and Immunity, Department of Microbiology and Immunology, Department of Pathology, and World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, Texas, USA
| | - Christl A Donnelly
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
- Department of Statistics, University of Oxford, Oxford, UK
| | - Amy Y Vittor
- Division of Infectious Disease and Global Medicine, Department of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
34
|
Fox JM, Huang L, Tahan S, Powell LA, Crowe JE, Wang D, Diamond MS. A cross-reactive antibody protects against Ross River virus musculoskeletal disease despite rapid neutralization escape in mice. PLoS Pathog 2020; 16:e1008743. [PMID: 32760128 PMCID: PMC7433899 DOI: 10.1371/journal.ppat.1008743] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 08/18/2020] [Accepted: 06/25/2020] [Indexed: 01/01/2023] Open
Abstract
Arthritogenic alphaviruses cause debilitating musculoskeletal disease and historically have circulated in distinct regions. With the global spread of chikungunya virus (CHIKV), there now is more geographic overlap, which could result in heterologous immunity affecting natural infection or vaccination. Here, we evaluated the capacity of a cross-reactive anti-CHIKV monoclonal antibody (CHK-265) to protect against disease caused by the distantly related alphavirus, Ross River virus (RRV). Although CHK-265 only moderately neutralizes RRV infection in cell culture, it limited clinical disease in mice independently of Fc effector function activity. Despite this protective phenotype, RRV escaped from CHK-265 neutralization in vivo, with resistant variants retaining pathogenic potential. Near the inoculation site, CHK-265 reduced viral burden in a type I interferon signaling-dependent manner and limited immune cell infiltration into musculoskeletal tissue. In a parallel set of experiments, purified human CHIKV immune IgG also weakly neutralized RRV, yet when transferred to mice, resulted in improved clinical outcome during RRV infection despite the emergence of resistant viruses. Overall, this study suggests that weakly cross-neutralizing antibodies can protect against heterologous alphavirus disease, even if neutralization escape occurs, through an early viral control program that tempers inflammation. The induction of broadly neutralizing antibodies is a goal of many antiviral vaccine programs. In this study, we show that cross-reactive monoclonal and polyclonal antibodies developed after CHIKV infection or immunization with relatively weak cross-neutralizing activity can protect against RRV-induced musculoskeletal disease in mice. Even though RRV rapidly escaped from neutralization, antibody therapy reduced inflammation in musculoskeletal tissues and decreased viral burden near the site of infection in a manner that required type I interferon signaling. These studies in mice show that broadly reactive antibodies with limited neutralizing activity still can confer protection against heterologous alphaviruses.
Collapse
Affiliation(s)
- Julie M. Fox
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Ling Huang
- MacroGenics, Rockville, Maryland, United States of America
| | - Stephen Tahan
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Laura A. Powell
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - James E. Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Vaccine Center and Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - David Wang
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Michael S. Diamond
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, United States of America
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
35
|
Guth S, Hanley KA, Althouse BM, Boots M. Ecological processes underlying the emergence of novel enzootic cycles: Arboviruses in the neotropics as a case study. PLoS Negl Trop Dis 2020; 14:e0008338. [PMID: 32790670 PMCID: PMC7425862 DOI: 10.1371/journal.pntd.0008338] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pathogens originating from wildlife (zoonoses) pose a significant public health burden, comprising the majority of emerging infectious diseases. Efforts to control and prevent zoonotic disease have traditionally focused on animal-to-human transmission, or "spillover." However, in the modern era, increasing international mobility and commerce facilitate the spread of infected humans, nonhuman animals (hereafter animals), and their products worldwide, thereby increasing the risk that zoonoses will be introduced to new geographic areas. Imported zoonoses can potentially "spill back" to infect local wildlife-a danger magnified by urbanization and other anthropogenic pressures that increase contacts between human and wildlife populations. In this way, humans can function as vectors, dispersing zoonoses from their ancestral enzootic systems to establish reservoirs elsewhere in novel animal host populations. Once established, these enzootic cycles are largely unassailable by standard control measures and have the potential to feed human epidemics. Understanding when and why translocated zoonoses establish novel enzootic cycles requires disentangling ecologically complex and stochastic interactions between the zoonosis, the human population, and the natural ecosystem. In this Review, we address this challenge by delineating potential ecological mechanisms affecting each stage of enzootic establishment-wildlife exposure, enzootic infection, and persistence-applying existing ecological concepts from epidemiology, invasion biology, and population ecology. We ground our discussion in the neotropics, where four arthropod-borne viruses (arboviruses) of zoonotic origin-yellow fever, dengue, chikungunya, and Zika viruses-have separately been introduced into the human population. This paper is a step towards developing a framework for predicting and preventing novel enzootic cycles in the face of zoonotic translocations.
Collapse
Affiliation(s)
- Sarah Guth
- Department of Integrative Biology, University of California, Berkeley, California, United States of America
| | - Kathryn A. Hanley
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
| | - Benjamin M. Althouse
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
- Epidemiology, Institute for Disease Modeling, Bellevue, Washington, United States of America
- Information School, University of Washington, Seattle, Washington, United States of America
| | - Mike Boots
- Department of Integrative Biology, University of California, Berkeley, California, United States of America
| |
Collapse
|
36
|
Azar SR, Campos RK, Bergren NA, Camargos VN, Rossi SL. Epidemic Alphaviruses: Ecology, Emergence and Outbreaks. Microorganisms 2020; 8:E1167. [PMID: 32752150 PMCID: PMC7464724 DOI: 10.3390/microorganisms8081167] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/23/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
Over the past century, the emergence/reemergence of arthropod-borne zoonotic agents has been a growing public health concern. In particular, agents from the genus Alphavirus pose a significant risk to both animal and human health. Human alphaviral disease presents with either arthritogenic or encephalitic manifestations and is associated with significant morbidity and/or mortality. Unfortunately, there are presently no vaccines or antiviral measures approved for human use. The present review examines the ecology, epidemiology, disease, past outbreaks, and potential to cause contemporary outbreaks for several alphavirus pathogens.
Collapse
Affiliation(s)
- Sasha R. Azar
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555-0609, USA;
| | - Rafael K. Campos
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX 77555-0609, USA;
| | | | - Vidyleison N. Camargos
- Host-Microorganism Interaction Lab, Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Shannan L. Rossi
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555-0609, USA;
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX 77555-0610, USA
| |
Collapse
|
37
|
Julia da Silva Pessoa Vieira C, José Ferreira da Silva D, Rigotti Kubiszeski J, Ceschini Machado L, Pena LJ, Vieira de Morais Bronzoni R, da Luz Wallau G. The Emergence of Chikungunya ECSA Lineage in a Mayaro Endemic Region on the Southern Border of the Amazon Forest. Trop Med Infect Dis 2020; 5:E105. [PMID: 32604785 PMCID: PMC7345197 DOI: 10.3390/tropicalmed5020105] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 12/13/2022] Open
Abstract
Anthropic changes on the edges of the tropical forests may facilitate the emergence of new viruses from the sylvatic environment and the simultaneous circulation of sylvatic and urban viruses in the human population. In this study, we investigated the presence of arboviruses (arthropod-borne viruses) in the sera of 354 patients, sampled from February 2014 to October 2018 in Sinop city. We sequenced the complete genomes of one chikungunya virus (CHIKV)-positive and one out of the 33 Mayaro virus (MAYV)-positive samples. The CHIKV genome obtained here belongs to the East/Central/South African (ECSA) genotype and the MAYV genome belongs to the L genotype. These genomes clustered with other viral strains from different Brazilian states, but the CHIKV strain circulating in Sinop did not cluster with other genomes from the Mato Grosso state, suggesting that at least two independent introductions of this virus occurred in Mato Grosso. Interestingly, the arrival of CHIKV in Sinop seems to not have caused a surge in human cases in the following years, as observed in the rest of the state, suggesting that cross immunity from MAYV infection might be protecting the population from CHIKV infection. These findings reinforce the need for continued genomic surveillance in order to evaluate how simultaneously circulating alphaviruses infecting the human population will unfold.
Collapse
Affiliation(s)
- Carla Julia da Silva Pessoa Vieira
- Health Sciences Institute, Federal University of Mato Grosso, Sinop 78550-728, MT, Brazil; (C.J.d.S.P.V.); (D.J.F.d.S.); (J.R.K.); (R.V.d.M.B.)
| | - David José Ferreira da Silva
- Health Sciences Institute, Federal University of Mato Grosso, Sinop 78550-728, MT, Brazil; (C.J.d.S.P.V.); (D.J.F.d.S.); (J.R.K.); (R.V.d.M.B.)
| | - Janaína Rigotti Kubiszeski
- Health Sciences Institute, Federal University of Mato Grosso, Sinop 78550-728, MT, Brazil; (C.J.d.S.P.V.); (D.J.F.d.S.); (J.R.K.); (R.V.d.M.B.)
| | - Laís Ceschini Machado
- Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife 50670-420, PE, Brazil; (L.C.M.); (L.J.P.)
| | - Lindomar José Pena
- Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife 50670-420, PE, Brazil; (L.C.M.); (L.J.P.)
| | - Roberta Vieira de Morais Bronzoni
- Health Sciences Institute, Federal University of Mato Grosso, Sinop 78550-728, MT, Brazil; (C.J.d.S.P.V.); (D.J.F.d.S.); (J.R.K.); (R.V.d.M.B.)
| | - Gabriel da Luz Wallau
- Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife 50670-420, PE, Brazil; (L.C.M.); (L.J.P.)
| |
Collapse
|
38
|
Nguyen W, Nakayama E, Yan K, Tang B, Le TT, Liu L, Cooper TH, Hayball JD, Faddy HM, Warrilow D, Allcock RJN, Hobson-Peters J, Hall RA, Rawle DJ, Lutzky VP, Young P, Oliveira NM, Hartel G, Howley PM, Prow NA, Suhrbier A. Arthritogenic Alphavirus Vaccines: Serogrouping Versus Cross-Protection in Mouse Models. Vaccines (Basel) 2020; 8:vaccines8020209. [PMID: 32380760 PMCID: PMC7349283 DOI: 10.3390/vaccines8020209] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 12/12/2022] Open
Abstract
Chikungunya virus (CHIKV), Ross River virus (RRV), o’nyong nyong virus (ONNV), Mayaro virus (MAYV) and Getah virus (GETV) represent arthritogenic alphaviruses belonging to the Semliki Forest virus antigenic complex. Antibodies raised against one of these viruses can cross-react with other serogroup members, suggesting that, for instance, a CHIKV vaccine (deemed commercially viable) might provide cross-protection against antigenically related alphaviruses. Herein we use human alphavirus isolates (including a new human RRV isolate) and wild-type mice to explore whether infection with one virus leads to cross-protection against viremia after challenge with other members of the antigenic complex. Persistently infected Rag1-/- mice were also used to assess the cross-protective capacity of convalescent CHIKV serum. We also assessed the ability of a recombinant poxvirus-based CHIKV vaccine and a commercially available formalin-fixed, whole-virus GETV vaccine to induce cross-protective responses. Although cross-protection and/or cross-reactivity were clearly evident, they were not universal and were often suboptimal. Even for the more closely related viruses (e.g., CHIKV and ONNV, or RRV and GETV), vaccine-mediated neutralization and/or protection against the intended homologous target was significantly more effective than cross-neutralization and/or cross-protection against the heterologous virus. Effective vaccine-mediated cross-protection would thus likely require a higher dose and/or more vaccinations, which is likely to be unattractive to regulators and vaccine manufacturers.
Collapse
Affiliation(s)
- Wilson Nguyen
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane 4029, Australia; (W.N.); (E.N.); (K.Y.); (B.T.); (T.T.L.); (D.J.R.); (V.P.L.)
| | - Eri Nakayama
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane 4029, Australia; (W.N.); (E.N.); (K.Y.); (B.T.); (T.T.L.); (D.J.R.); (V.P.L.)
- Department of Virology I, National Institute of Infectious Diseases, Tokyo 162-0052, Japan
| | - Kexin Yan
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane 4029, Australia; (W.N.); (E.N.); (K.Y.); (B.T.); (T.T.L.); (D.J.R.); (V.P.L.)
| | - Bing Tang
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane 4029, Australia; (W.N.); (E.N.); (K.Y.); (B.T.); (T.T.L.); (D.J.R.); (V.P.L.)
| | - Thuy T. Le
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane 4029, Australia; (W.N.); (E.N.); (K.Y.); (B.T.); (T.T.L.); (D.J.R.); (V.P.L.)
| | - Liang Liu
- Experimental Therapeutics Laboratory, School of Pharmacy & Medical Sciences, University of South Australia Cancer Research Institute, SA 5000, Australia; (L.L.); (T.H.C.); (J.D.H.)
| | - Tamara H. Cooper
- Experimental Therapeutics Laboratory, School of Pharmacy & Medical Sciences, University of South Australia Cancer Research Institute, SA 5000, Australia; (L.L.); (T.H.C.); (J.D.H.)
| | - John D. Hayball
- Experimental Therapeutics Laboratory, School of Pharmacy & Medical Sciences, University of South Australia Cancer Research Institute, SA 5000, Australia; (L.L.); (T.H.C.); (J.D.H.)
| | - Helen M. Faddy
- Research and Development Laboratory, Australian Red Cross Lifeblood, Kelvin Grove, Qld 4059, Australia;
| | - David Warrilow
- Public Health Virology Laboratory, Queensland Health Forensic and Scientific Services, PO Box 594, Archerfield, Qld 4108, Australia;
| | - Richard J. N. Allcock
- School of Biomedical Sciences, University of Western Australia, Crawley 6009, Australia;
| | - Jody Hobson-Peters
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Qld 4072, Australia; (J.H.-P.); (R.A.H.); (P.Y.)
| | - Roy A. Hall
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Qld 4072, Australia; (J.H.-P.); (R.A.H.); (P.Y.)
- Australian Infectious Disease Research Centre, Brisbane, Qld 4027 & 4072, Australia
| | - Daniel J. Rawle
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane 4029, Australia; (W.N.); (E.N.); (K.Y.); (B.T.); (T.T.L.); (D.J.R.); (V.P.L.)
| | - Viviana P. Lutzky
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane 4029, Australia; (W.N.); (E.N.); (K.Y.); (B.T.); (T.T.L.); (D.J.R.); (V.P.L.)
| | - Paul Young
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, Qld 4072, Australia; (J.H.-P.); (R.A.H.); (P.Y.)
- Australian Infectious Disease Research Centre, Brisbane, Qld 4027 & 4072, Australia
| | - Nidia M. Oliveira
- Deptartment of Microbiology, University of Western Australia, Perth, WA 6009, Australia;
| | - Gunter Hartel
- Statistics Unit, QIMR Berghofer Medical Research Institute, Brisbane, Qld 4029, Australia;
| | | | - Natalie A. Prow
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane 4029, Australia; (W.N.); (E.N.); (K.Y.); (B.T.); (T.T.L.); (D.J.R.); (V.P.L.)
- Experimental Therapeutics Laboratory, School of Pharmacy & Medical Sciences, University of South Australia Cancer Research Institute, SA 5000, Australia; (L.L.); (T.H.C.); (J.D.H.)
- Australian Infectious Disease Research Centre, Brisbane, Qld 4027 & 4072, Australia
- Correspondence: (N.A.P.); (A.S.)
| | - Andreas Suhrbier
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane 4029, Australia; (W.N.); (E.N.); (K.Y.); (B.T.); (T.T.L.); (D.J.R.); (V.P.L.)
- Australian Infectious Disease Research Centre, Brisbane, Qld 4027 & 4072, Australia
- Correspondence: (N.A.P.); (A.S.)
| |
Collapse
|
39
|
In-depth characterization of a novel live-attenuated Mayaro virus vaccine candidate using an immunocompetent mouse model of Mayaro disease. Sci Rep 2020; 10:5306. [PMID: 32210270 PMCID: PMC7093544 DOI: 10.1038/s41598-020-62084-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/24/2020] [Indexed: 02/06/2023] Open
Abstract
Mayaro virus (MAYV) is endemic in South American countries where it is responsible for sporadic outbreaks of acute febrile illness. The hallmark of MAYV infection is a highly debilitating and chronic arthralgia. Although MAYV emergence is a potential threat, there are no specific therapies or licensed vaccine. In this study, we developed a murine model of MAYV infection that emulates many of the most relevant clinical features of the infection in humans and tested a live-attenuated MAYV vaccine candidate (MAYV/IRES). Intraplantar inoculation of a WT strain of MAYV into immunocompetent mice induced persistent hypernociception, transient viral replication in target organs, systemic production of inflammatory cytokines, chemokines and specific humoral IgM and IgG responses. Inoculation of MAYV/IRES in BALB/c mice induced strong specific cellular and humoral responses. Moreover, MAYV/IRES vaccination of immunocompetent and interferon receptor-defective mice resulted in protection from disease induced by the virulent wt MAYV strain. Thus, this study describes a novel model of MAYV infection in immunocompetent mice and highlights the potential role of a live-attenuated MAYV vaccine candidate in host's protection from disease induced by a virulent MAYV strain.
Collapse
|